1
|
Alvarado-Ocampo J, Abrahams-Sandí E, Retana-Moreira L. Overview of extracellular vesicles in pathogens with special focus on human extracellular protozoan parasites. Mem Inst Oswaldo Cruz 2024; 119:e240073. [PMID: 39319874 PMCID: PMC11421424 DOI: 10.1590/0074-02760240073] [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: 04/01/2024] [Accepted: 07/09/2024] [Indexed: 09/26/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid-bilayered membrane-delimited particles secreted by almost any cell type, involved in different functions according to the cell of origin and its state. From these, cell to cell communication, pathogen-host interactions and modulation of the immune response have been widely studied. Moreover, these vesicles could be employed for diagnostic and therapeutic purposes, including infections produced by pathogens of diverse types; regarding parasites, the secretion, characterisation, and roles of EVs have been studied in particular cases. Moreover, the heterogeneity of EVs presents challenges at every stage of studies, which motivates research in this area. In this review, we summarise some aspects related to the secretion and roles of EVs from several groups of pathogens, with special focus on the most recent research regarding EVs secreted by extracellular protozoan parasites.
Collapse
Affiliation(s)
- Johan Alvarado-Ocampo
- Universidad de Costa Rica, Facultad de Microbiología, Centro de Investigación en Enfermedades Tropicales, San José, Costa Rica
| | - Elizabeth Abrahams-Sandí
- Universidad de Costa Rica, Facultad de Microbiología, Centro de Investigación en Enfermedades Tropicales, San José, Costa Rica
- Universidad de Costa Rica, Facultad de Microbiología, Departamento de Parasitología, San José, Costa Rica
| | - Lissette Retana-Moreira
- Universidad de Costa Rica, Facultad de Microbiología, Centro de Investigación en Enfermedades Tropicales, San José, Costa Rica
- Universidad de Costa Rica, Facultad de Microbiología, Departamento de Parasitología, San José, Costa Rica
| |
Collapse
|
2
|
Vidal AS, Zauli RC, Batista WL, Xander P. Extracellular vesicles release from protozoa parasite and animal model. CURRENT TOPICS IN MEMBRANES 2024; 94:85-106. [PMID: 39370214 DOI: 10.1016/bs.ctm.2024.06.007] [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: 10/08/2024]
Abstract
Diseases caused by protozoan parasites, such as leishmaniasis, trypanosomiasis, and malaria, are highly complex and together continue to cause high annual morbidity and mortality. The search for new compounds in environmental biodiversity, repositioning known drugs, and developing vaccines using old and innovative technologies have been employed to discover vaccines and new and alternative treatments. Extracellular vesicles (EVs) can carry parasite antigens, creating a new possibility to develop an effective and affordable platform for treatment, vaccines, and drug delivery. Thus, the evaluation of EVs in animal models can and should be explored among the countless biomedical applications. Herein, we will address the concept of EVs, their acquisition and characterization in protozoan parasite models, and the primary studies using these vesicles in therapeutic applications.
Collapse
Affiliation(s)
- Andrey Sladkevicius Vidal
- Programa de Pós-Graduação Biologia-Química, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema, Brazil
| | - Rogéria Cristina Zauli
- Programa de Pós-Graduação Biologia-Química, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema, Brazil
| | - Wagner Luiz Batista
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema, Brazil
| | - Patricia Xander
- Programa de Pós-Graduação Biologia-Química, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema, Brazil; Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema, Brazil.
| |
Collapse
|
3
|
Yang D, Liu Y, Ren Y, Hao L, Zhang X, Chen H, Liu J. Giardia intestinalis extracellular vesicles induce changes in gene expression in human intestinal epithelial cells in vitro. Exp Parasitol 2024; 262:108788. [PMID: 38759775 DOI: 10.1016/j.exppara.2024.108788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Giardiasis is a common waterborne zoonotic disease caused by Giardia intestinalis. Upon infection, Giardia releases excretory and secretory products (ESPs) including secreted proteins (SPs) and extracellular vesicles (EVs). Although the interplay between ESPs and intestinal epithelial cells (IECs) has been previously described, the functions of EVs in these interactions and their differences from those of SPs require further exploration. In the present study, EVs and EV-depleted SPs were isolated from Giardia ESPs. Proteomic analyses of isolated SPs and EVs showed 146 and 91 proteins, respectively. Certain unique and enriched proteins have been identified in SPs and EVs. Transcriptome analysis of Caco-2 cells exposed to EVs showed 96 differentially expressed genes (DEGs), with 56 upregulated and 40 downregulated genes. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) indicated that Caco-2 genes related to metabolic processes, the HIF-1 signaling pathway, and the cAMP signaling pathway were affected. This study provides new insights into host-parasite interactions, highlighting the potential significance of EVs on IECs during infections.
Collapse
Affiliation(s)
- Dongming Yang
- Shanghai Veterinary Research Institute, Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China; College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan 610041, China
| | - Yingnan Liu
- Shanghai Veterinary Research Institute, Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Yupeng Ren
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan 610041, China
| | - Lili Hao
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan 610041, China
| | - Xichen Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Hongjun Chen
- Shanghai Veterinary Research Institute, Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| | - Jingyi Liu
- Shanghai Veterinary Research Institute, Biosafety Research Center, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| |
Collapse
|
4
|
Zheng G, Sun S, Zhang G, Liang X. miR-144 affects the immune response and activation of inflammatory responses in Cynoglossus semilaevis by regulating the expression of CsMAPK6. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109578. [PMID: 38670413 DOI: 10.1016/j.fsi.2024.109578] [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: 01/29/2024] [Revised: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
MicroRNAs are increasingly recognized for their pivotal role in the immune system, yet the specific regulatory functions of fish-derived microRNAs remain largely unexplored. In this research, we discovered a novel miRNA, Cse-miR-144, in the Chinese tongue sole (Cynoglossus semilaevis), characterized by a 73-base pair precursor and a 21-nucleotide mature sequence. Our findings revealed that the expression of Cse-miR-144 was notably inhibited by various Vibrio species. Utilizing bioinformatics and dual-luciferase assay techniques, we established that the pro-inflammatory cytokine gene CsMAPK6 is a direct target of Cse-miR-144. Subsequent in vitro and in vivo western blotting analyses confirmed that Cse-miR-144 can effectively reduce the protein levels of CsMAPK6 post-transcriptionally. Moreover, CsMAPK6 is known to be involved in the activation of the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-kB). Additional investigations using qPCR and ELISA demonstrated that suppression of Cse-miR-144 leads to an upsurge in the liver mRNA levels of various immune genes (including MYD88, TRAF6, NF-κB, TRAF2, TRAF3, and TNF), alongside a marked increase in the production and secretion of pro-inflammatory cytokines (IL-1β, IL-6, and IL-8) in the bloodstream of C. semilaevis. These findings collectively underscore the potential of Cse-miR-144 as a key inhibitor of CsMAPK and its crucial role in modulating the immune and inflammatory responses in teleost fish. Compared to the siRNA, miRNA is a better tool in controlling the expression of target gene with a lower cost.
Collapse
Affiliation(s)
- Guiliang Zheng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Siqi Sun
- Biodesign Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Guosong Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China.
| | - Xia Liang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China.
| |
Collapse
|
5
|
Kwaku GN, Ward RA, Vyas JM, Harding HB. Host innate immune systems gather intel on invading microbes via pathogen-derived extracellular vesicles. EXTRACELLULAR VESICLE 2024; 3:100043. [PMID: 38939756 PMCID: PMC11209872 DOI: 10.1016/j.vesic.2024.100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles released into the extracellular milieu from various cell types including host cells and pathogens that infect them. As carriers of nucleic acids, proteins, lipids, metabolites, and virulence factors, EVs act as delivery vehicles for intercellular communication and quorum sensing. Innate immune cells have the capacity to intercept, internalize, and interpret 'messages' contained within these EVs. This review categorizes the ability of EVs secreted by bacterial, parasitic, and fungal pathogens to trigger both pro- and anti-inflammatory innate immune responses in the host. Understanding molecular pathways and inflammatory responses activated in innate immune cells upon pathogen-derived EV stimulation is critical to gain insight into potential therapeutics and combat these infectious diseases.
Collapse
Affiliation(s)
- Geneva N. Kwaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca A. Ward
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jatin M. Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hannah Brown Harding
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
6
|
Zhang K, Zhang M, Su H, Zhao F, Wang D, Zhang Y, Cao G, Zhang Y. Regulation of Inflammatory Responses of Cow Mammary Epithelial Cells through MAPK Signaling Pathways of IL-17A Cytokines. Animals (Basel) 2024; 14:1572. [PMID: 38891619 PMCID: PMC11171030 DOI: 10.3390/ani14111572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The aim of this study is to explore the mechanism of IL-17A infection in the development of bacterial mastitis in dairy cows. In this study, RT-qPCR and ELISA were used to measure the promoting effect of IL-17A on the generation of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and chemokine (IL-8). In addition, Western blot (WB) was applied to measure the influences of IL-17A on the inflammation-related ERK and p38 proteins in the MAPK pathways. The results show that under the stimulation of LPS on cow mammary epithelial cells (CMECs), cytokines IL-1β, IL-6, IL-8, TNF-α, and IL-17A will exhibit significantly increased expression levels (p < 0.05). With inhibited endogenous expression of IL-17A, cytokines IL-1β, IL-6, IL-8, and TNF-α will present reduced genetic expression (p < 0.01), with reduced phosphorylation levels of ERK and p38 proteins in the MAPK signaling pathways (p < 0.001). Upon the exogenous addition of the IL-17A cytokine, the genetic expression of cytokines IL-1β, IL-6, IL-8, and TNF-α will increase (p < 0.05), with increased phosphorylation levels of the ERK and p38 proteins in the MAPK signaling pathways (p < 0.001). These results indicate that under the stimulation of CMECs with LPS, IL-17A can be expressed together with relevant inflammatory cytokines. Meanwhile, the inflammatory responses of mammary epithelial cells are directly proportional to the expression levels of IL-17A inhibited alone or exogenously added. In summary, this study shows that IL-17A could be used as an important indicator for assessing the bacterial infections of mammary glands, indicating that IL-17A could be regarded as one potential therapeutic target for mastitis.
Collapse
Affiliation(s)
- Kai Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Min Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Hong Su
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Feifei Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Daqing Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Guifang Cao
- College of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Yong Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| |
Collapse
|
7
|
Rodríguez-González J, Wilkins-Rodríguez AA, Gutiérrez-Kobeh L. Human Dendritic Cell Maturation Is Modulated by Leishmania mexicana through Akt Signaling Pathway. Trop Med Infect Dis 2024; 9:118. [PMID: 38787051 PMCID: PMC11126033 DOI: 10.3390/tropicalmed9050118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Dendritic cells (DC) along with macrophages are the main host cells of the intracellular parasite Leishmania. DC traverse a process of maturation, passing through an immature state with phagocytic ability to a mature one where they can modulate the immune response through the secretion of cytokines. Several studies have demonstrated that Leishmania inhibits DC maturation. Nevertheless, when cells are subjected to a second stimulus such as LPS/IFN-γ, they manage to mature. In the maturation process of DC, several signaling pathways have been implicated, importantly MAPK. On the other hand, Akt is a signaling pathway deeply involved in cell survival. Some Leishmania species have shown to activate MAPK and Akt in different cells. The aim of this work was to investigate the role of ERK and Akt in the maturation of monocyte-derived DC (moDC) infected with L. mexicana. moDC were infected with L. mexicana metacyclic promastigotes, and the phosphorylation of ERK and Akt, the expression of MHCII and CD86 and IL-12 transcript, and secretion were determined in the presence or absence of an Akt inhibitor. We showed that L. mexicana induces a sustained Akt and ERK phosphorylation, while the Akt inhibitor inhibits it. Moreover, the infection of moDC downregulates CD86 expression but not MHCII, and the Akt inhibitor reestablishes CD86 expression and 12p40 production. Thus, L. mexicana can modulate DC maturation though Akt signaling.
Collapse
Affiliation(s)
- Jorge Rodríguez-González
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez, Oaxaca C.P. 68120, Mexico;
| | - Arturo A. Wilkins-Rodríguez
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico;
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico;
| |
Collapse
|
8
|
Menezes SA, Tasca T. Extracellular vesicles in parasitic diseases - from pathogenesis to future diagnostic tools. Microbes Infect 2024; 26:105310. [PMID: 38316376 DOI: 10.1016/j.micinf.2024.105310] [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: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Parasitic diseases are still a major public health problem especially among individuals of low socioeconomic status in underdeveloped countries. In recent years it has been demonstrated that parasites can release extracellular vesicles that participate in the host-parasite communication, immune evasion, and in governing processes associated with host infection. Extracellular vesicles are membrane-bound structures released into the extracellular space that can carry several types of biomolecules, including proteins, lipids, nucleic acids, and metabolites, which directly impact the target cells. Extracellular vesicles have attracted wide attention due to their relevance in host-parasite communication and for their potential value in applications such as in the diagnostic biomarker discovery. This review of the literature aimed to join the current knowledge on the role of extracellular vesicles in host-parasite interaction and summarize its molecular content, providing information for the acquisition of new tools that can be used in the diagnosis of parasitic diseases. These findings shed light to the potential of extracellular vesicle cargo derived from protozoan parasites as novel diagnostic tools.
Collapse
Affiliation(s)
- Saulo Almeida Menezes
- Faculdade de Farmácia e Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 90610-000, RS, Brazil.
| | - Tiana Tasca
- Faculdade de Farmácia e Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre 90610-000, RS, Brazil.
| |
Collapse
|
9
|
Chai F, Peng H, Qin L, Liu C, Zeng Y, Wang R, Xu G, Wang R, Wei G, Huang H, Lan Y, Chen W, Wang C. MicroRNA miR-181d-5p regulates the MAPK signaling pathway by targeting mitogen-activated protein kinase 8 (MAPK8) to improve lupus nephritis. Gene 2024; 893:147961. [PMID: 37931853 DOI: 10.1016/j.gene.2023.147961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Lupus nephritis (LN) is a common immune disease. The microRNA (miR)-181d-5p is a potential target for treating kidney injury. However, the therapeutic role of miR-181d-5p in LN has not been investigated. This study aimed to investigate the role of miR-181d-5p in targeting mitogen-activated protein kinase 8 (MAPK8) and stimulating the MAPK signaling pathway in LN. METHODS RT-qPCR was performed to identify the variations in miR-181d-5p expression in peripheral blood mononuclear cells (PBMCs) obtained from 42 LN patients, 30 healthy individuals, 6 MRL/lpr mice and 6 C57BL/6 mice. Western blot was used to detect the effect of miR-181d-5p on the MAPK signaling pathway in THP-1 cells and MRL/lpr mice. Enzyme-linked immunosorbent assay (ELISA) was utilized to detect the effect of miR-181d-5p on antinuclear antibodies and inflammatory factors. A dual-luciferase reporter assay was used to verify whether miR-181d-5p directly targets MAPK8. Flow cytometry was performed to evaluate apoptosis rates in transfected THP-1 cells. RESULTS miR-181d-5p expression was downregulated in PBMCs of LN patients (P < 0.01) and MRL/lpr mice (P < 0.05). A dual luciferase reporter assay demonstrated that miR-181d-5p inhibits MAPK8 (P < 0.01). Overexpression of miR-181d-5p inhibited the phosphorylation of p38 (P < 0.001) and p44/42 (P < 0.01). Moreover, miR-181d-5p decreased the apoptosis rate of THP-1 cells (P < 0.001), and reduced the secretion of IL-6 (P < 0.01) and TNF-α (P < 0.01). Furthermore, overexpression of miR-181d-5p decreased anti-dsDNA antibody (P < 0.05), anti-Sm antibody (P < 0.01), and fibrosis levels in MRL/lpr mice. CONCLUSION Upregulation of miR-181d-5p showed anti-inflammatory and anti-apoptotic effects on THP-1 cells in vitro and kidney injury in vivo. These effects were achieved by miR-181d-5p targeting MAPK8 to inhibit phosphorylation of p38 and p44/42. These results may offer new insights for improving therapeutic strategies against lupus nephritis.
Collapse
Affiliation(s)
- Fu Chai
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Huixin Peng
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Linxiu Qin
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Chunhong Liu
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yonglong Zeng
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Rong Wang
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Guidan Xu
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Rongqi Wang
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Guijiang Wei
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Huayi Huang
- Roswell Park Comprehensive Cancer Center, Surgical Oncology, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Yan Lan
- Department of Dermatology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Wencheng Chen
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China.
| | - Chunfang Wang
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China.
| |
Collapse
|
10
|
Saeedi P, Eslami G, Tohidfar M, Jafari-Nodushan A, Vakili M. Differential gene expression (DGE) analysis in persons with a history of giardiasis. AMB Express 2024; 14:1. [PMID: 38170269 PMCID: PMC10764694 DOI: 10.1186/s13568-023-01657-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
Giardiasis, which is caused by Giardia duodenalis, has clinical symptoms such as steatorrhea and can be very dangerous in children. In addition, some documents reported that this parasite is present inside the tissue of patients with cancer. In this study, we analyzed the gene expression profiles of some main genes important to apoptosis and anti-apoptosis in humans.Expression profile arrays of Genomic Spatial Event (GSE) 113666, GSE113667, and GSE113679 obtained from Gene Expression Omnibus were used for meta-analysis using R commands. Cytoscape and STRING databases used the protein-protein Interaction network. Then, the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analysis was performed. Similar genes in Homo sapiens were identified using Basic Local Alignment Search Tool analysis. The validation was performed on eight people using real-time Polymerase chain reaction. In addition to the candidate genes, the gene expression of some other genes, including Serine/Threonine Kinase 1 (AKT1), Cyclin Dependent Kinase Inhibitor 2A (CDKN2A), Kirsten Rat Sarcoma (KRAS), and Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha (PIK3CA) were also examined. Analysis of the expression of serum amyloid A1 (SAA1), Regenerating Islet-Derived 3 Gamma (REG3G), and REG3A genes did not show any difference between the two groups of healthy and diseased people. Examining the mean expression of the four genes AKT1, CDKN2A, KRAS, and PIK3CA showed that three genes of AKT1, CDKN2A, and KRAS had increased expression in people with a history of giardiasis compared to healthy people. We showed that the gene expression pattern differs in apoptosis and anti-apoptosis signaling in people with a history of giardiasis. Giardia duodenalis seems to induce post-non-infectious symptoms with stimulation of human gene expression.
Collapse
Affiliation(s)
- Parnia Saeedi
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Gilda Eslami
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Masoud Tohidfar
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - AbbasAli Jafari-Nodushan
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahmood Vakili
- Department of Community and Preventive Medicine, Health Monitoring Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
11
|
Sun M, Zhao Z, Li Y, Cao L, Li J, Zhang X, Li X, Zhang N, Cheng S, Wang X, Gong P. Giardia VSPAS7 protein attenuates Giardia intestinalis-induced host macrophage pyroptosis. Parasit Vectors 2023; 16:359. [PMID: 37821972 PMCID: PMC10566177 DOI: 10.1186/s13071-023-05949-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/27/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The unicellular protozoan parasite Giardia intestinalis, which primarily infects humans and animals such as cattle and sheep, is having a major negative impact on public health. Giardia is able to evade the recognition and elimination of the host immune system because of the trophozoite surface and extracellular vesicles (EVs) covered by variant-specific surface proteins (VSPs). As key proteins for immune evasion, whether VSPs can regulate Giardia-induced pyroptosis and promote Giardia evasion of host immune responses has not been reported. METHODS To examine the role of Giardia VSPAS7 on Giardia-induced activation of the signaling pathway, secretion of pro-inflammatory cytokines, pyroptosis and the mechanism involved, we constructed the pcDNA3.1-vspas7 expression plasmid and transfected this plasmid into mouse macrophages. Key proteins for pyroptosis, IL-1β secretion and LDH release were detected in pcDNA3.1-vspas7-transfected wild-type (WT) cells and NLRP3-deficient cells by western blot, ELISA and LDH assays, respectively. The interactions of Giardia VSPAS7 and mouse NLRP3 were examined using immunofluorescence assays (IFA), co-immunoprecipitation (Co-IP) and bimolecular fluorescence complementation (BiFC) assays. RESULTS VSPAS7 could decrease the levels of phosphorylated-p65 (P-p65), P-IκBα and P-ERK caused by Giardia and reduce the production levels of Giardia-induced pro-inflammatory cytokine IL-6, IL-12 p40 and TNF-α. The results showed that VSPAS7 inhibited Giardia-mediated activation of NF-κB, ERK/MAPK signaling and secretion of pro-inflammatory cytokines. Furthermore, VSPAS7 suppressed Giardia-induced macrophage pyroptosis by reducing GSDMD cleavage, caspase-1 activation, IL-1β secretion and LDH release. We further found that VSPAS7 could interact with mouse NLRP3 directly, and in NLRP3-deficient cells the suppression of Giardia-induced macrophage pyroptosis by VSPAS7 was significantly attenuated. CONCLUSIONS Overall, VSPAS7 could inhibit Giardia-induced activation of signaling pathways and pyroptosis in host macrophages, allowing Giardia evasion of host immune responses. Studies on Giardia VSP-mediated immune evasion provide an important theoretical basis for in-depth studies on Giardia pathogenicity.
Collapse
Affiliation(s)
- Min Sun
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Zhiteng Zhao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Ying Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Lili Cao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Jianhua Li
- Jilin Academy of Animal Husbandry and Veterinary Medicine, Changchun, 130062 China
| | - Xichen Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Xin Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Nan Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Shuqin Cheng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Xiaocen Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Pengtao Gong
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| |
Collapse
|
12
|
Sharma M, Lozano-Amado D, Chowdhury D, Singh U. Extracellular Vesicles and Their Impact on the Biology of Protozoan Parasites. Trop Med Infect Dis 2023; 8:448. [PMID: 37755909 PMCID: PMC10537256 DOI: 10.3390/tropicalmed8090448] [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: 08/07/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-membrane-bound structures produced naturally by all cells and have a variety of functions. EVs act as vehicles for transporting important molecular signals from one cell to another. Several parasites have been shown to secrete EVs, and their biological functions have been extensively studied. EVs have been shown to facilitate communication with the host cells (such as modulation of the host's immune system or promoting attachment and invasion into the host cells) or for communication between parasitic cells (e.g., transferring drug-resistance genes or factors modulating stage conversion). It is clear that EVs play an important role in host-parasite interactions. In this review, we summarized the latest research on the EVs secreted by protozoan parasites and their role in host-parasite and parasite-parasite communications.
Collapse
Affiliation(s)
- Manu Sharma
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.S.); (D.L.-A.); (D.C.)
| | - Daniela Lozano-Amado
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.S.); (D.L.-A.); (D.C.)
| | - Debabrata Chowdhury
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.S.); (D.L.-A.); (D.C.)
| | - Upinder Singh
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.S.); (D.L.-A.); (D.C.)
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
13
|
DeMichele E, Sosnowski O, Buret AG, Allain T. Regulatory Functions of Hypoxia in Host-Parasite Interactions: A Focus on Enteric, Tissue, and Blood Protozoa. Microorganisms 2023; 11:1598. [PMID: 37375100 PMCID: PMC10303274 DOI: 10.3390/microorganisms11061598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Body tissues are subjected to various oxygenic gradients and fluctuations and hence can become transiently hypoxic. Hypoxia-inducible factor (HIF) is the master transcriptional regulator of the cellular hypoxic response and is capable of modulating cellular metabolism, immune responses, epithelial barrier integrity, and local microbiota. Recent reports have characterized the hypoxic response to various infections. However, little is known about the role of HIF activation in the context of protozoan parasitic infections. Growing evidence suggests that tissue and blood protozoa can activate HIF and subsequent HIF target genes in the host, helping or hindering their pathogenicity. In the gut, enteric protozoa are adapted to steep longitudinal and radial oxygen gradients to complete their life cycle, yet the role of HIF during these protozoan infections remains unclear. This review focuses on the hypoxic response to protozoa and its role in the pathophysiology of parasitic infections. We also discuss how hypoxia modulates host immune responses in the context of protozoan infections.
Collapse
Affiliation(s)
- Emily DeMichele
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
14
|
Papadopoulos DK, Lattos A, Giantsis IA, Theodorou JA, Michaelidis B, Feidantsis K. The impact of ascidian biofouling on the farmed Mediterranean mussel Mytilus galloprovincialis physiology and welfare, revealed by stress biomarkers. BIOFOULING 2023:1-18. [PMID: 37144608 DOI: 10.1080/08927014.2023.2209015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
In biofouling communities, ascidians are among the most damaging species, presenting severe threats, such as depressed growth rates and decreased chances of lower survival, to shellfish aquaculture. However, little is known concerning the fouled shellfish physiology. In an effort to obtain information for the magnitude of stress caused by ascidians to farmed Mytilus galloprovincialis, five seasonal samplings took place in a mussel aquaculture farm suffering from ascidian biofoulants, in Vistonicos Bay, Greece. The dominant ascidian species were recorded and several stress biomarkers, including Hsp gene expression at both mRNA and protein levels, as well as MAPKs levels, and enzymatic activities of intermediate metabolism were examined. Almost all investigated biomarkers revealed elevated stress levels in fouled mussels compared to non-fouled. This enhanced physiological stress seems to be season-independent and can be attributed to the oxidative stress and/or feed deprivation caused by ascidian biofouling, thus illuminating the biological impact of this phenomenon.
Collapse
Affiliation(s)
- Dimitrios K Papadopoulos
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Lattos
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis A Giantsis
- Department of Animal Science, Faculty of Agricultural Sciences, University of Western Macedonia, Florina, Greece
| | - John A Theodorou
- Department of Fisheries & Aquaculture, University of Patras, Mesolonghi, Greece
| | - Basile Michaelidis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Feidantsis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Zhao P, Li J, Li X, Dong J, Wang X, Zhang N, Li S, Sun M, Zhang X, Wang Z, Liang M, Li Y, Cao L, Gong P. The NLRP3 inflammasome recognizes alpha-2 and alpha-7.3 giardins and decreases the pathogenicity of Giardia duodenalis in mice. Parasit Vectors 2023; 16:85. [PMID: 36869360 PMCID: PMC9983531 DOI: 10.1186/s13071-023-05688-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/01/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Giardia duodenalis is a parasitic organism that can cause giardiasis, an intestinal infection, particularly prevalent in young children, with clinical symptoms of diarrhea. We previously reported that extracellular G. duodenalis triggers intracellular nucleotide-binding oligomerization-like receptor 3 (NLRP3) inflammasome activation and regulates the host inflammatory response by secreting extracellular vesicles (EVs). However, the exact pathogen-associated molecular patterns in G. duodenalis EVs (GEVs) involved in this process and the role of the NLRP3 inflammasome in giardiasis remain to be elucidated. METHODS Recombinant eukaryotic expression plasmids of pcDNA3.1(+)-alpha-2 and alpha-7.3 giardins in GEVs were constructed, transfected into primary mouse peritoneal macrophages and screened by measuring the expression levels of the inflammasome target molecule caspase-1 p20. The preliminary identification of G. duodenalis alpha-2 and alpha-7.3 giardins was further verified by measuring the protein expression levels of key molecules of the NLRP3 inflammasome (NLRP3, pro-interleukin-1 beta [IL-1β], pro-caspase-1, and caspase-1 p20), the secretion levels of IL-1β, the level of apoptosis speck-like protein (ASC) oligomerization and the immunofluorescence localization of NLRP3 and ASC. The roles of the NLRP3 inflammasome in G. duodenalis pathogenicity were then evaluated using mice in which NLRP3 activation was blocked (NLRP3-blocked mice), and body weight, parasite burden in the duodenum and histopathological changes in the duodenum were monitored. In addition, we explored whether alpha-2 and alpha-7.3 giardins triggered IL-1β secretion in vivo through the NLRP3 inflammasome and determined the roles of these molecules in G. duodenalis pathogenicity in mice. RESULTS Alpha-2 and alpha-7.3 giardins triggered NLRP3 inflammasome activation in vitro. This led to caspase-1 p20 activation, upregulation of the protein expression levels of NLRP3, pro-IL-1β and pro-caspase-1, significant enhancement of IL-1β secretion, ASC speck formation in the cytoplasm and also induction of ASC oligomerization. Deletion of the NLRP3 inflammasome aggravated G. duodenalis pathogenicity in mice. Compared to wild-type mice gavaged with cysts, mice gavaged with cysts in NLRP3-blocked mice displayed increased trophozoite loads and severe duodenal villus damage, characterized by necrotic crypts with atrophy and branching. In vivo assays revealed that alpha-2 and alpha-7.3 giardins could induce IL-1β secretion through the NLRP3 inflammasome and that immunization with alpha-2 and alpha-7.3 giardins decreased G. duodenalis pathogenicity in mice. CONCLUSIONS Overall, the results of the present study revealed that alpha-2 and alpha-7.3 giardins trigger host NLRP3 inflammasome activation and decrease G. duodenalis infection ability in mice, which are promising targets for the prevention of giardiasis.
Collapse
Affiliation(s)
- Panpan Zhao
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Jianhua Li
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Xin Li
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Jingquan Dong
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China.,Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, Jiangsu Province, People's Republic of China
| | - Xiaocen Wang
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Nan Zhang
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Shan Li
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Min Sun
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Xichen Zhang
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Zhibang Wang
- College of Life Science, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Min Liang
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Ying Li
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China
| | - Lili Cao
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China. .,Jilin Academy of Animal Husbandry and Veterinary Medicine, Changchun, 130062, Jilin Province, People's Republic of China.
| | - Pengtao Gong
- State Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin Province, People's Republic of China.
| |
Collapse
|
16
|
Ferreira B, Lourenço Á, Sousa MDC. Protozoa-Derived Extracellular Vesicles on Intercellular Communication with Special Emphasis on Giardia lamblia. Microorganisms 2022; 10:microorganisms10122422. [PMID: 36557675 PMCID: PMC9788250 DOI: 10.3390/microorganisms10122422] [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: 10/08/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Parasitic diseases are an important worldwide problem threatening human health and affect millions of people. Acute diarrhea, intestinal bleeding, malabsorption of nutrients and nutritional deficiency are some of the issues related to intestinal parasitic infections. Parasites are experts in subvert the host immune system through different kinds of mechanisms. There are evidences that extracellular vesicles (EVs) have an important role in dissemination of the disease and in modulating the host immune system. Released by almost all types of cells, these nanovesicles are a natural secretory product containing multiple components of interest. The EVs are classified as apoptotic bodies, microvesicles, exosomes, ectosomes, and microparticles, according to their physical characteristics, biochemical composition and cell of origin. Interestingly, EVs play an important role in intercellular communication between parasites as well as with the host cells. Concerning Giardia lamblia, it is known that this parasite release EVs during it life cycle that modulate the parasite growth and adherence as well the immune system of the host. Here we review the recently updates on protozoa EVs, with particular emphasis on the role of EVs released by the flagellate protozoa G. lamblia in cellular communication and its potential for future applications as vaccine, therapeutic agent, drug delivery system and as diagnostic or prognostic biomarker.
Collapse
Affiliation(s)
- Bárbara Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIVG-Vasco da Gama Research Center, EUVG-Vasco da Gama University School, 3020-210 Coimbra, Portugal
| | - Ágata Lourenço
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Maria do Céu Sousa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
17
|
Grajeda BI, De Chatterjee A, Villalobos CM, Pence BC, Ellis CC, Enriquez V, Roy S, Roychowdhury S, Neumann AK, Almeida IC, Patterson SE, Das S. Giardial lipid rafts share virulence factors with secreted vesicles and participate in parasitic infection in mice. Front Cell Infect Microbiol 2022; 12:974200. [PMID: 36081774 PMCID: PMC9445159 DOI: 10.3389/fcimb.2022.974200] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Giardia lamblia, a protozoan parasite, is a major cause of waterborne infection, worldwide. While the trophozoite form of this parasite induces pathological symptoms in the gut, the cyst form transmits the infection. Since Giardia is a noninvasive parasite, the actual mechanism by which it causes disease remains elusive. We have previously reported that Giardia assembles cholesterol and GM1 glycosphingolipid-enriched lipid rafts (LRs) that participate in encystation and cyst production. To further delineate the role of LRs in pathogenesis, we isolated LRs from Giardia and subjected them to proteomic analysis. Various cellular proteins including potential virulence factors-e.g., giardins, variant surface proteins, arginine deaminases, elongation factors, ornithine carbomyltransferases, and high cysteine-rich membrane proteins-were found to be present in LRs. Since Giardia secretes virulence factors encapsulated in extracellular vesicles (EVs) that induce proinflammatory responses in hosts, EVs released by the parasite were isolated and subjected to nanoparticle tracking and proteomic analysis. Two types of EV-i.e., small vesicles (SVs; <100 nm, exosome-like particles) and large vesicles (LVs; 100-400 nm, microvesicle-like particles)-were identified and found to contain a diverse group of proteins including above potential virulence factors. Although pretreatment of the parasite with two giardial lipid raft (gLR) disruptors, nystatin (27 μM) and oseltamivir (20 μM), altered the expression profiles of virulence factors in LVs and SVs, the effects were more robust in the case of SVs. To examine the potential role of rafts and vesicles in pathogenicity, Giardia-infected mice were treated with oseltamivir (1.5 and 3.0 mg/kg), and the shedding of cysts were monitored. We observed that this drug significantly reduced the parasite load in mice. Taken together, our results suggest that virulence factors partitioning in gLRs, released into the extracellular milieu via SVs and LVs, participate in spread of giardiasis and could be targeted for future drug development.
Collapse
Affiliation(s)
- Brian I. Grajeda
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Atasi De Chatterjee
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Carmen M. Villalobos
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Breanna C. Pence
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Cameron C. Ellis
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Vanessa Enriquez
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Sourav Roy
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Sukla Roychowdhury
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Aaron K. Neumann
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Igor C. Almeida
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Steven E. Patterson
- Center for Drug Design, University of Minnesota, Minneapolis, MN, United States
| | - Siddhartha Das
- Infectious Disease and Immunology, Border Biomedical Research Center and the Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
18
|
Wang X, Chen J, Zheng J. The state of the art of extracellular vesicle research in protozoan infection. Front Genet 2022; 13:941561. [PMID: 36035188 PMCID: PMC9417467 DOI: 10.3389/fgene.2022.941561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Protozoan diseases seriously affect the health of human beings, livestock and poultry and lead to high economic and medical costs. Extracellular vesicles (EVs) are membranous structures formed through biological processes that play important roles in immune regulation. Studies have shown that parasites transmit information to hosts through EVs to modulate host immune responses. The major roles played by EVs released from parasites involve facilitating parasitization of the host. In this review, we discuss relevant recently obtained data on EVs secreted by different kinds of protozoa, including their molecular mechanisms, and discuss the roles played by EVs in the occurrence and development of parasitic diseases.
Collapse
Affiliation(s)
- Xinlei Wang
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Jie Chen
- Institute of Theoretical Chemistry, Jilin University, Changchun, China
| | - Jingtong Zheng
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, China
- *Correspondence: Jingtong Zheng,
| |
Collapse
|
19
|
Gong Z, Zhang S, Gu B, Cao J, Mao W, Yao Y, Zhao J, Ren P, Zhang K, Liu B. Codonopsis pilosula polysaccharide attenuates Escherichia coli-induced acute lung injury in mice. Food Funct 2022; 13:7999-8011. [DOI: 10.1039/d2fo01221a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Acute lung injury (ALI) is an inflammatory lung disease could be caused by bacterial infection. Lipopolysaccharide (LPS), a prototype pathogen-associated molecular pattern (PAMP) from gram-negative bacteria such as Escherichia coli...
Collapse
|
20
|
Liu L, Yang Y, Fang R, Zhu W, Wu J, Li X, Patankar JV, Li W. Giardia duodenalis and Its Secreted PPIB Trigger Inflammasome Activation and Pyroptosis in Macrophages through TLR4-Induced ROS Signaling and A20-Mediated NLRP3 Deubiquitination. Cells 2021; 10:cells10123425. [PMID: 34943932 PMCID: PMC8700504 DOI: 10.3390/cells10123425] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/18/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
The extracellular protozoan parasite Giardia duodenalis is a well-known and important causative agent of diarrhea on a global scale. Macrophage pyroptosis has been recognized as an important innate immune effector mechanism against intracellular pathogens. Yet, the effects of noninvasive Giardia infection on macrophage pyroptosis and the associated molecular triggers and regulators remain poorly defined. Here we initially observed that NLRP3 inflammasome-mediated pyroptosis was activated in Giardia-treated macrophages, and inhibition of ROS, NLRP3, or caspase-1 could block GSDMD cleavage, IL-1β, IL-18 and LDH release, and the cell viability reduction. We also confirmed that Giardia-induced NLRP3 inflammasome activation was involved in its K63 deubiquitination. Thus, six candidate deubiquitinases were screened, among which A20 was identified as an effective regulator. We then screened TLRs on macrophage membranes and found that upon stimulation TLR4 was tightly correlated to ROS enhancement, A20-mediated NLRP3 deubiquitination, and pyroptotic signaling. In addition, several Giardia-secreted proteins were predicted as trigger factors via secretome analysis, of which peptidyl-prolyl cis-trans isomerase B (PPIB) independently induced macrophage pyroptosis. This was similar to the findings from the trophozoite treatment, and also led to the TLR4-mediated activation of NLRP3 through K63 deubiquitination by A20. Collectively, the results of this study have significant implications for expanding our understanding of host defense mechanisms after infection with G. duodenalis.
Collapse
Affiliation(s)
- Lin Liu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
| | - Yongwu Yang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
| | - Rui Fang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
| | - Weining Zhu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
| | - Jingxue Wu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
| | - Xiaoyun Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
| | - Jay V. Patankar
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany;
| | - Wei Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (L.L.); (Y.Y.); (R.F.); (W.Z.); (J.W.); (X.L.)
- Correspondence: or
| |
Collapse
|