1
|
Wu F, Chen Y, Chen X, Tong D, Zhou J, Du Z, Yao C, Yang Y, Du A, Ma G. Nematode serine protease inhibitor SPI-I8 negatively regulates host NF-κB signalling by hijacking MKRN1-mediated polyubiquitination of RACK1. Commun Biol 2025; 8:356. [PMID: 40032982 PMCID: PMC11876351 DOI: 10.1038/s42003-025-07803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
Parasitic roundworms are remarkable for their ability to manipulate host immune systems and ameliorate inflammatory diseases. Although much is known about the nature of nematode effectors in immune modulation, little is known about the action mode of these molecules. Here, we report that a serine protease inhibitor SPI-I8 in the extracellular vesicles of blood-feeding nematodes like Ancylostoma ceylanicum, Haemonchus contortus and Nippostrongylus brasiliensis, effectively halts excessive inflammatory responses in vitro and in vivo. We demonstrate that H. contortus SPI-I8 promotes the role of a negative regulator of RACK1 and enhances the effects of RACK1 on tumor necrosis factor (TNF)-α-IκB kinases (IKKs)-nuclear factor kappa beta (NF-κB) axis in mammalian cells, by hijacking E3 ubiquitin protein ligase MKRN1-mediated polyubiquitination of RACK1. Administration of recombinant N. brasiliensis SPI-I8 effectively protects mice from dextran sulfate sodium (DSS)-induced colitis and lipopolysaccharide (LPS)-induced sepsis. Considering the structural and functional conservation of SPI-I8s among Strongylida nematodes and the conservation of interactive mediators (i.e., MKRN1 and RACK1) among mammals, our findings provide insights into the host-parasite interface where parasitic roundworms secret molecules to suppress host inflammatory responses. Harnessing these findings should underpin the exploitation of nematode's immunomodulators to relief excessive inflammation associated diseases in animals and humans.
Collapse
Affiliation(s)
- Fei Wu
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Yanqiong Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueqiu Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Danni Tong
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingru Zhou
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Frontier Science Center for Brain and Brain-machine integration, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhendong Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chaoqun Yao
- Ross University School of Veterinary Medicine and One Health Center for Zoonoses and Tropical Veterinary Medicine, Ross University School of Veterinary Medicine, Basseterre, St. Kitts, Trinidad and Tobago
| | - Yi Yang
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Aifang Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangxu Ma
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Xinchang, China.
| |
Collapse
|
2
|
Cheng CJ, Wang LC, Chu LJ, Chen KY, Huang CY, Lan KL, Huang KY. Extracellular vesicles from fifth-stage larval Angiostrongylus cantonensis upregulate cholesterol biosynthesis and suppress NLRP2-associated inflammatory responses in mouse astrocytes. mSystems 2025; 10:e0101424. [PMID: 39636121 PMCID: PMC11748502 DOI: 10.1128/msystems.01014-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
Angiostrongylus cantonensis is a zoonotic parasite that causes severe symptoms in humans, including eosinophilic meningitis and eosinophilic meningoencephalitis. Extracellular vesicles (EVs) derived from helminthes have been implicated in regulating host survival and immune response. However, the roles of A. cantonensis EVs in modulating parasite pathogenesis and host immune response remain poorly understood. Herein, we characterized EVs derived from A. cantonensis fifth-stage larvae (L5) and adult worms. Ultrastructural features showed that EVs from adult worms are smaller in size compared with those from L5. Proteomic analysis identified stage-specific proteins packaged in L5 and adult worm EVs. To investigate the crosstalk between L5 EVs and host cells, RNA sequencing analysis was conducted to identify the differentially expressed genes (DEGs) and enriched biological pathways in mouse astrocytes treated with L5 EVs. GO and KEGG enrichment analysis demonstrated that the pathways related to "cholesterol biosynthesis" are significantly upregulated in L5 EV-treated astrocytes. Based on the transcriptomic data, we observed a downregulated trend of NOD-like receptors (NLRs) protein 2 (NLRP2), a key regulator of brain inflammation, in mouse astrocytes treated with L5 EVs. To validate this result, we utilized ATP to induce the expression of NLRP2 inflammasome-related genes and proteins, as well as the secretion of downstream cytokines. Notably, ATP-induced overexpression of NLRP2 inflammasome-related molecules was significantly reduced in mouse astrocytes upon L5 EV treatment. Collectively, our data suggest that A. cantonensis L5 EVs enhance cholesterol synthesis and potentially modulate immune response by reducing NLRP2 inflammasome-related signaling in non-permissive host cells.IMPORTANCEAngiostrongylus cantonensis is a significant causative agent of eosinophilic meningitis and eosinophilic meningoencephalitis in humans. Helminth-derived extracellular vesicles (EVs) are known to play a crucial role in parasite pathogenesis and host immunomodulation. However, the protein compositions of A. cantonensis EVs and their roles in parasite pathogenesis and host immune response remain unclear. Our results demonstrate for the first time the distinct protein compositions in A. cantonensis L5 and adult worm EVs. The highly abundant proteins in L5 EVs that have immunomodulatory or pathogenic potential in the host deserve further investigation. Additionally, the uptake of L5 EVs by mouse astrocytes significantly upregulates cholesterol synthesis and suppresses ATP-induced NLRP2 inflammasome-related signaling. This study highlights the immunomodulatory roles of L5 EVs in non-permissive hosts, suggesting their potential as therapeutic targets and vaccine candidates against A. cantonensis.
Collapse
Affiliation(s)
- Chien-Ju Cheng
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, Taiwan
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Lian-Chen Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Lichieh Julie Chu
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
- Department of Otolaryngology—Head & Neck Surgery, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Kuang-Yao Chen
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Ching-Yun Huang
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei City, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City, Taiwan
| | - Kuo-Lun Lan
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Kuo-Yang Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, Taiwan
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
3
|
Barrero-Torres DM, Herrera-Torres G, Pérez J, Martínez-Moreno Á, Martínez-Moreno FJ, Flores-Velázquez LM, Buffoni L, Rufino-Moya PJ, Ruiz-Campillo MT, Molina-Hernández V. Unraveling the microRNAs Involved in Fasciolosis: Master Regulators of the Host-Parasite Crosstalk. Int J Mol Sci 2024; 26:204. [PMID: 39796061 PMCID: PMC11719827 DOI: 10.3390/ijms26010204] [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: 11/22/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Fasciolosis is a neglected tropical disease caused by helminth parasites of the genus Fasciola spp., including Fasciola hepatica (F. hepatica) and Fasciola gigantica (F. gigantica), being a major zoonotic problem of human and animal health. Its control with antihelminthics is becoming ineffective due to the increase in parasite resistance. Developing new therapeutic protocols is crucial to a deeper knowledge of the molecular bases in the host-parasite interactions. The high-throughput omics technologies have dramatically provided unprecedented insights into the complexity of the molecular host-parasite crosstalk. MicroRNAs (miRNAs) are key players as critical regulators in numerous biological processes, modifying the gene expression of cells by degradation of messenger RNA (mRNA), regulating transcription and translation functions, protein positioning, cell cycle integrity, differentiation and apoptosis. The large-scale exploration of miRNAs, including the miRNome, has offered great scientific knowledge of steps in fasciolosis, further scrutinizing the pathogenesis, the growth and development of their strains and their interaction with the host for the survival of the different parasite stages. This review compiles the updated knowledge related to miRNAs involved in fasciolosis and the generated miRNome, highlighting the importance of these key molecules in the host-parasite interactions and the pathogenesis of Fasciola spp. directing towards the development of new biotherapeutic protocols for the control of fasciolosis.
Collapse
Affiliation(s)
- Diana María Barrero-Torres
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (D.M.B.-T.); (G.H.-T.); (J.P.); (L.M.F.-V.)
| | - Guillem Herrera-Torres
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (D.M.B.-T.); (G.H.-T.); (J.P.); (L.M.F.-V.)
| | - José Pérez
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (D.M.B.-T.); (G.H.-T.); (J.P.); (L.M.F.-V.)
| | - Álvaro Martínez-Moreno
- Departamento de Sanidad Animal (Área de Parasitología), UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (Á.M.-M.); (F.J.M.-M.); (L.B.); (P.J.R.-M.)
| | - Francisco Javier Martínez-Moreno
- Departamento de Sanidad Animal (Área de Parasitología), UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (Á.M.-M.); (F.J.M.-M.); (L.B.); (P.J.R.-M.)
| | - Luis Miguel Flores-Velázquez
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (D.M.B.-T.); (G.H.-T.); (J.P.); (L.M.F.-V.)
- Unidad de Anatomía, Histología y Patología Veterinaria, Escuela de Medicina Veterinaria, Facultad de Ciencias Naturales, Universidad San Sebastián, Campus Puerto Montt, Puerto Montt 5480000, Chile
| | - Leandro Buffoni
- Departamento de Sanidad Animal (Área de Parasitología), UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (Á.M.-M.); (F.J.M.-M.); (L.B.); (P.J.R.-M.)
| | - Pablo José Rufino-Moya
- Departamento de Sanidad Animal (Área de Parasitología), UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (Á.M.-M.); (F.J.M.-M.); (L.B.); (P.J.R.-M.)
| | - María Teresa Ruiz-Campillo
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (D.M.B.-T.); (G.H.-T.); (J.P.); (L.M.F.-V.)
| | - Verónica Molina-Hernández
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), Universidad de Córdoba, Campus de Rabanales, Edificio Sanidad Animal, 14071 Córdoba, Spain; (D.M.B.-T.); (G.H.-T.); (J.P.); (L.M.F.-V.)
| |
Collapse
|
4
|
Xu S, Wang D, Tan L, Lu J. The role of NLRP3 inflammasome in type 2 inflammation related diseases. Autoimmunity 2024; 57:2310269. [PMID: 38332696 DOI: 10.1080/08916934.2024.2310269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
Type 2 inflammation related diseases, such as atopic dermatitis, asthma, and allergic rhinitis, are diverse and affect multiple systems in the human body. It is common for individuals to have multiple co-existing type 2 inflammation related diseases, which can impose a significant financial and living burden on patients. However, the exact pathogenesis of these diseases is still unclear. The NLRP3 inflammasome is a protein complex composed of the NLRP3 protein, ASC, and Caspase-1, and is activated through various mechanisms, including the NF-κB pathway, ion channels, and lysosomal damage. The NLRP3 inflammasome plays a role in the immune response to pathogens and cellular damage. Recent studies have indicated a strong correlation between the abnormal activation of NLRP3 inflammasome and the onset of type 2 inflammation. Additionally, it has been demonstrated that suppressing NLRP3 expression effectively diminishes the inflammatory response, highlighting its promising therapeutic applications. Therefore, this article reviews the role of NLRP3 inflammasome in the development and therapy of multiple type 2 inflammation related diseases.
Collapse
Affiliation(s)
- Shenming Xu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Lina Tan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Jianyun Lu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
5
|
Flores-Sotelo I, Juárez N, González MI, Chávez A, Vannan DT, Eksteen B, Terrazas LI, Reyes JL. Endogenous innate sensor NLRP3 is a key component in peritoneal macrophage dynamics required for cestode establishment. Immunol Res 2024; 72:948-963. [PMID: 38842647 PMCID: PMC11564225 DOI: 10.1007/s12026-024-09496-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
The NLRP3 receptor can assemble inflammasome platforms to trigger inflammatory responses; however, accumulating evidence suggests that it can also display anti-inflammatory properties. Here, we explored the role of nucleotide-binding oligomerization domain pyrin-containing protein 3 (NLRP3) in Taenia crassiceps experimental infection, which requires immune polarization into a Th2-type profile and peritoneal influx of suppressive macrophages for successful colonization. NLRP3 deficient mice (NLRP3-/-) were highly resistant against T. crassiceps, relative to wild-type (WT) mice. Resistance in NLRP3-/- mice was associated with a diminished IL-4 output, high levels of IL-15, growth factor for both innate and adaptive lymphocytes, and a dramatic decrease in peritoneum-infiltrating suppressive macrophages. Also, a transcriptional analysis on bone marrow-derived macrophages exposed to Taenia-secreted antigens and IL-4 revealed that NLRP3-/- macrophages express reduced transcripts of relm-α and PD-1 ligands, markers of alternative activation and suppressive ability, respectively. Finally, we found that the resistance displayed by NLRP3-/- mice is transferred through intestinal microbiota exchange, since WT mice co-housed with NLRP3-/- mice were significantly more resistant than WT animals preserving their native microbiota. Altogether, these data demonstrate that NLRP3 is a component of innate immunity required for T. crassiceps to establish, most likely contributing to macrophage recruitment, and controlling lymphocyte-stimulating cytokines such as IL-15.
Collapse
Affiliation(s)
- Irán Flores-Sotelo
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Natalia Juárez
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Marisol I González
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Auraamellaly Chávez
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Danielle T Vannan
- Boston Scientific Corporation, Urology Division, 200 Boston Scientific Way, Marlborough, MA, USA
| | | | - Luis I Terrazas
- Laboratorio de Inmunoparasitología, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - José L Reyes
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico.
| |
Collapse
|
6
|
Rizk SKA, Gaber A, Hamdy H, Labeeb A, Fouda E, Hosny A, El Derbaly SA. Concurrent genotyping and expression of NLRP3 inflammasome in pityriasis versicolor patient's skin lesions. Arch Dermatol Res 2024; 316:501. [PMID: 39093484 DOI: 10.1007/s00403-024-03221-8] [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: 05/30/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024]
Abstract
The goal of this study is to investigate the impact of the rs35829419 SNP on the serum level of NLRP3, and to assess the relationship between NLRP3 and its SNP and vulnerability to Pityriasis versicolor. Pityriasis versicolor (PV) is one of the most frequent skin conditions linked to skin pigmentation changes. Malassezia plays a key role in the pathogenesis of PV. A case-control study, 50 patients with pityriasis versicolor and 44 healthy controls. Real-time PCR was used to genotype NLRP3 (rs35829419) and ELISA assay of NLRP3 levels in tissue samples. There was a significantly higher median NLPR3 levels in PV patients than controls. A significant predominance of A allele of Q 705 K was in patients than controls. The risk of having the disease in the presence of A allele is nearly 10 times than having C allele. In PV patients, there was a significant relationship between NLPR3 levels and Q 705 K genotypes with higher NLPR3 levels in AA genotype. A potential correlation between PV and the Q705K polymorphism, pointing to evidence of NLRP3 alteration in PV patients. The NLRP3 inflammasome may be an appropriate therapeutic target for Malassezia-associated skin disorders.
Collapse
Affiliation(s)
- Sara Kamal AbdelMaksoud Rizk
- Medical Biochemistry and Molecular Biology Department, Faculty of medicine, Menoufia University, Shebin Elkom, Egypt.
| | - Azza Gaber
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | - Hadir Hamdy
- Dermatology and Andrology Department in Dekernes General Hospital, Dakahlia Governorate, el dakahleya, Egypt
| | - Azza Labeeb
- Microbiology and immunology Department, Faculty of medicine, Menoufia University, Shebin Elkom, Egypt
| | - Eman Fouda
- Department of Chemistry, Biochemistry Division, Faculty of Science, Menoufia University, Shebin Elkom, Egypt
| | - Ammar Hosny
- Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Sara A El Derbaly
- Medical Biochemistry and Molecular Biology Department, Faculty of medicine, Menoufia University, Shebin Elkom, Egypt
| |
Collapse
|
7
|
Alipour S, Mardi A, Shajari N, Kazemi T, Sadeghi MR, Ahmadian Heris J, Masoumi J, Baradaran B. Unmasking the NLRP3 inflammasome in dendritic cells as a potential therapeutic target for autoimmunity, cancer, and infectious conditions. Life Sci 2024; 348:122686. [PMID: 38710282 DOI: 10.1016/j.lfs.2024.122686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.
Collapse
Affiliation(s)
- Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Tsankov BK, Luchak A, Carr C, Philpott DJ. The effects of NOD-like receptors on adaptive immune responses. Biomed J 2024; 47:100637. [PMID: 37541620 PMCID: PMC10796267 DOI: 10.1016/j.bj.2023.100637] [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: 04/28/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023] Open
Abstract
It has long been appreciated that cues from the innate immune system orchestrate downstream adaptive immune responses. Although previous work has focused on the roles of Toll-like receptors in this regard, relatively little is known about how Nod-like receptors instruct adaptive immunity. Here we review the functions of different members of the Nod-like receptor family in orchestrating effector and anamnestic adaptive immune responses. In particular, we address the ways in which inflammasome and non-inflammasome members of this family affect adaptive immunity under various infectious and environmental contexts. Furthermore, we identify several key mechanistic questions that studies in this field have left unaddressed. Our aim is to provide a framework through which immunologists in the adaptive immune field may view their questions through an innate-immune lens and vice-versa.
Collapse
Affiliation(s)
- Boyan K Tsankov
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Alexander Luchak
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Charles Carr
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Sais D, Chowdhury S, Dalton JP, Tran N, Donnelly S. Both host and parasite non-coding RNAs co-ordinate the regulation of macrophage gene expression to reduce pro-inflammatory immune responses and promote tissue repair pathways during infection with fasciola hepatica. RNA Biol 2024; 21:62-77. [PMID: 39344634 PMCID: PMC11445894 DOI: 10.1080/15476286.2024.2408706] [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] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Parasitic worms (helminths) establish chronic infection within mammalian hosts by strategically regulating their host's immune responses. Deciphering the mechanisms by which host non-coding RNAs (ncRNA) co-ordinate the activation and regulation of immune cells is essential to understanding host immunity and immune-related pathology. It is also important to comprehend how pathogens secrete specific ncRNAs to manipulate gene expression of host immune cells and influence their response to infection. To investigate the contribution of both host and helminth derived ncRNAs to the activation and/or regulation of innate immune responses during a parasite infection, we examined ncRNA expression in the peritoneal macrophages from mice infected with Fasciola hepatica. We discovered the presence of several parasitic-derived miRNAs within host macrophages at 6 hrs and 18 hrs post infection. Target prediction analysis showed that these Fasciola miRNAs regulate host genes associated with the activation of host pro-inflammatory macrophages. Concomitantly, there was a distinct shift in host ncRNA expression, which was significant at 5 days post-infection. Prediction analysis suggested that these host ncRNAs target a different cohort of host genes compared to the parasite miRNAs, although the functional outcome was predicted to be similar i.e. reduced pro-inflammatory response and the promotion of a reparative/tolerant phenotype. Taken together, these observations uncover the interplay between host and parasitic ncRNAs and reveal a complementary regulation of the immune response that allows the parasite to evade immune detection and promote tissue repair for the host. These findings will provide a new understanding of the molecular interaction between parasites and host.
Collapse
Affiliation(s)
- Dayna Sais
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Sumaiya Chowdhury
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Technology Sydney, Ultimo, NSW, Australia
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - John. P. Dalton
- Molecular Parasitology Laboratory, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Nham Tran
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
- Molecular Parasitology Laboratory, School of Natural Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
10
|
Lekki-Jóźwiak J, Bąska P. The Roles of Various Immune Cell Populations in Immune Response against Helminths. Int J Mol Sci 2023; 25:420. [PMID: 38203591 PMCID: PMC10778651 DOI: 10.3390/ijms25010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Helminths are multicellular parasites that are a substantial problem for both human and veterinary medicine. According to estimates, 1.5 billion people suffer from their infection, resulting in decreased life quality and burdens for healthcare systems. On the other hand, these infections may alleviate autoimmune diseases and allergy symptoms. The immune system is programmed to combat infections; nevertheless, its effector mechanisms may result in immunopathologies and exacerbate clinical symptoms. This review summarizes the role of the immune response against worms, with an emphasis on the Th2 response, which is a hallmark of helminth infections. We characterize non-immune cells (enteric tuft cells-ETCs) responsible for detecting parasites, as well as the role of hematopoietic-derived cells (macrophages, basophils, eosinophils, neutrophils, innate lymphoid cells group 2-ILC2s, mast cells, T cells, and B cells) in initiating and sustaining the immune response, as well as the functions they play in granulomas. The aim of this paper is to review the existing knowledge regarding the immune response against helminths, to attempt to decipher the interactions between cells engaged in the response, and to indicate the gaps in the current knowledge.
Collapse
Affiliation(s)
- Janina Lekki-Jóźwiak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
11
|
Liu J, Liu H, Liu H, Teng Y, Qin N, Ren X, Xia X. Live and pasteurized Akkermansia muciniphila decrease susceptibility to Salmonella Typhimurium infection in mice. J Adv Res 2023; 52:89-102. [PMID: 36996967 PMCID: PMC10555781 DOI: 10.1016/j.jare.2023.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/02/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
INTRODUCTION The gut microbiome is vital for providing resistance against colonized pathogenicbacteria. Recently, specific commensal species have become recognized as important mediators of host defense against microbial infection by a variety of mechanisms. OBJECTIVES To examine the contribution of live and pasteurized A. muciniphila to defend against the intestinal pathogen Salmonella Typhimurium in a streptomycin-treated mouse model of infection. METHODS C57B6J mice were pretreated with phosphate-buffered saline (PBS), live Akkermansia muciniphila (AKK), and pasteurized A. muciniphila (pAKK) for two weeks, then mice were infected by S. Typhimurium SL 1344. 16S rRNA-based gut microbiota analysis was performed before and after infection. Bacterial counts in feces and tissues, histopathological analysis, gut barrier-related gene expression, and antimicrobial peptides were examined. Co-housing was performed to examine the role of microbiota in the change of susceptibility of mice to infection. RESULTS AKK and pAKK markedly decreased Salmonella fecal and systemic burdens and reduced inflammation during infection. Notably, further characterization of AKK and pAKK protective mechanisms revealed different candidate protective pathways. AKK promoted gutbarrier gene expression and the secretion of antimicrobial peptides, and co-housing studies suggested that AKK-associated microbial community played a role in attenuating infection. Moreover, pAKK had a positive effect on NLRP3 in infected mice. We verified that pretreatment of pAKK could promote the expression of NLRP3, and enhance the antimicrobial activity of macrophage, likely through increasing the production of reactive oxygen (ROS), nitric oxide (NO), and inflammatory cytokines. CONCLUSION Our study demonstrates that live or pasteurized A. muciniphila can be effective preventive measures for alleviating S. Typhimurium-induced disease, highlighting the potential of developing Akkermansia-based probiotics or postbiotics for the prevention of Salmonellosis.
Collapse
Affiliation(s)
- Jiaxiu Liu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Hongli Liu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Huanhuan Liu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Yue Teng
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Ningbo Qin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaomeng Ren
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
12
|
Honda TSB, Ku J, Anders HJ. Cell type-specific roles of NLRP3, inflammasome-dependent and -independent, in host defense, sterile necroinflammation, tissue repair, and fibrosis. Front Immunol 2023; 14:1214289. [PMID: 37564649 PMCID: PMC10411525 DOI: 10.3389/fimmu.2023.1214289] [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: 04/29/2023] [Accepted: 06/27/2023] [Indexed: 08/12/2023] Open
Abstract
The NLRP3 inflammasome transforms a wide variety of infectious and non-infectious danger signals that activate pro-inflammatory caspases, which promote the secretion of IL-1β and IL-18, and pyroptosis, a pro-inflammatory form of cell necrosis. Most published evidence documents the presence and importance of the NLRP3 inflammasome in monocytes, macrophages, and neutrophils during host defense and sterile forms of inflammation. In contrast, in numerous unbiased data sets, NLRP3 inflammasome-related transcripts are absent in non-immune cells. However, an increasing number of studies report the presence and functionality of the NLRP3 inflammasome in almost every cell type. Here, we take a closer look at the reported cell type-specific expression of the NLRP3 inflammasome components, review the reported inflammasome-dependent and -independent functions, and discuss possible explanations for this discrepancy.
Collapse
Affiliation(s)
| | | | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| |
Collapse
|
13
|
Tran LS, Ying L, D'Costa K, Wray-McCann G, Kerr G, Le L, Allison CC, Ferrand J, Chaudhry H, Emery J, De Paoli A, Colon N, Creed S, Kaparakis-Liaskos M, Como J, Dowling JK, Johanesen PA, Kufer TA, Pedersen JS, Mansell A, Philpott DJ, Elgass KD, Abud HE, Nachbur U, Croker BA, Masters SL, Ferrero RL. NOD1 mediates interleukin-18 processing in epithelial cells responding to Helicobacter pylori infection in mice. Nat Commun 2023; 14:3804. [PMID: 37365163 DOI: 10.1038/s41467-023-39487-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
The interleukin-1 family members, IL-1β and IL-18, are processed into their biologically active forms by multi-protein complexes, known as inflammasomes. Although the inflammasome pathways that mediate IL-1β processing in myeloid cells have been defined, those involved in IL-18 processing, particularly in non-myeloid cells, are still not well understood. Here we report that the host defence molecule NOD1 regulates IL-18 processing in mouse epithelial cells in response to the mucosal pathogen, Helicobacter pylori. Specifically, NOD1 in epithelial cells mediates IL-18 processing and maturation via interactions with caspase-1, instead of the canonical inflammasome pathway involving RIPK2, NF-κB, NLRP3 and ASC. NOD1 activation and IL-18 then help maintain epithelial homoeostasis to mediate protection against pre-neoplastic changes induced by gastric H. pylori infection in vivo. Our findings thus demonstrate a function for NOD1 in epithelial cell production of bioactive IL-18 and protection against H. pylori-induced pathology.
Collapse
Affiliation(s)
- L S Tran
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, Australia
| | - L Ying
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, Australia
| | - K D'Costa
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - G Wray-McCann
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - G Kerr
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - L Le
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - C C Allison
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - J Ferrand
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - H Chaudhry
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - J Emery
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, Australia
| | - A De Paoli
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - N Colon
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - S Creed
- Monash Micro Imaging, Monash University, Melbourne, VIC, Australia
| | - M Kaparakis-Liaskos
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - J Como
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - J K Dowling
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - P A Johanesen
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - T A Kufer
- Department of Immunology, University of Hohenheim, Institute of Nutritional Medicine, Stuttgart, Germany
| | | | - A Mansell
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, Australia
| | - D J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - K D Elgass
- Monash Micro Imaging, Monash University, Melbourne, VIC, Australia
| | - H E Abud
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - U Nachbur
- Cell Signalling and Cell Death Division, WEHI, Melbourne, VIC, Australia
| | - B A Croker
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Inflammation Division, WEHI, Melbourne, VIC, Australia
| | - S L Masters
- Inflammation Division, WEHI, Melbourne, VIC, Australia
| | - R L Ferrero
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
- Inflammation Division, WEHI, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Xaplanteri P, Rodis N, Potsios C. Gut Microbiota Crosstalk with Resident Macrophages and Their Role in Invasive Amebic Colitis and Giardiasis-Review. Microorganisms 2023; 11:1203. [PMID: 37317178 DOI: 10.3390/microorganisms11051203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 06/16/2023] Open
Abstract
The innate immune response is highly dependent on the action of macrophages. They are abundant in the intestine subepithelial lamina propria of the mucosa, where they deploy multiple tasks and play a critical role. The balance between the gut microbiota and M2 macrophages is critical for gut health and homeostasis. Gut microbiota has the power to change macrophage phenotype and replenish the resident macrophage niche during and post infection. As far as the extracellular enteric parasitic infections invasive amebic colitis and giardiasis are concerned, a change of macrophages phenotype to a pro-inflammatory state is dependent on direct contact of the protozoan parasites with host cells. Macrophages induce strong pro-inflammatory response by inflammasome activation and secretion of interleukin IL-1β. Inflammasomes play a key role in the response to cellular stress and microbe attacks. The balance between gut mucosal homeostasis and infection is dependent on the crosstalk between microbiota and resident macrophages. Parasitic infections involve NLRP1 and NLRP3 inflammasome activation. For Entamoeba histolytica and Giardia duodenalis infections, inflammasome NLRP3 activation is crucial to promote the host defenses. More studies are needed to further elucidate possible therapeutic and protective strategies against these protozoan enteric parasites' invasive infections in humans.
Collapse
Affiliation(s)
- Panagiota Xaplanteri
- Department of Microbiology, General Hospital of Eastern Achaia, 25001 Kalavrita, Greece
| | - Nikiforos Rodis
- Department of Surgery, University General Hospital of Patras, 26332 Patras, Greece
| | - Charalampos Potsios
- Department of Internal Medicine, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
15
|
Pan TX, Huang HB, Lu HN, Zhao GX, Quan Y, Li JY, Xue Y, Zhu ZY, Wang Y, Shi CW, Wang N, Yang GL, Wang CF. NLRP3 Plays a Key Role in Antihelminth Immunity in the Enteral and Parenteral Stages of Trichinella spiralis-Infected Mice. Infect Immun 2023; 91:e0038222. [PMID: 36939354 PMCID: PMC10112270 DOI: 10.1128/iai.00382-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 02/28/2023] [Indexed: 03/21/2023] Open
Abstract
Trichinellosis is an important foodborne zoonosis, and no effective treatments are yet available. Nod-like receptor (NLR) plays a critical role in the host response against nematodes. Therefore, we aimed to explore the role of the NLRP3 inflammasome (NLRP3) during the adult, migrating, and encysted stages of Trichinella spiralis infection. The mice were treated with the specific NLRP3 inhibitor MCC950 after inoculation with T. spiralis. Then, the role that NLRP3 plays during T. spiralis infection of mice was evaluated using enzyme-linked immunosorbent assay (ELISA), Western blotting, flow cytometry, histopathological evaluation, bone marrow-derived macrophage (BMDM) stimulation, and immunofluorescence. The in vivo results showed that NLRP3 enhanced the Th1 immune response in the adult and migrating stages and weakened the Th2 immune response in the encysted stage. NLRP3 promoted the release of proinflammatory factors (interferon gamma [IFN-γ]) and suppressed the release of anti-inflammatory factors (interleukin 4 [IL-4]). Pathological changes were also improved in the absence of NLRP3 in mice during T. spiralis infection. Importantly, a significant reduction in adult worm burden and muscle larvae burden at 7 and 35 days postinfection was observed in mice treated with the specific NLRP3 inhibitor MCC950. In vitro, we first demonstrated that NLRP3 in macrophages can be activated by T. spiralis proteins and promotes IL-1β and IL-18 release. This study revealed that NLRP3 is involved in the host response to T. spiralis infection and that targeted inhibition of NLRP3 enhanced the Th2 response and accelerated T. spiralis expulsion. These findings may help in the development of protocols for controlling trichinellosis.
Collapse
Affiliation(s)
- Tian-Xu Pan
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hui-Nan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Guang-Xun Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yu Quan
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jun-Yi Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ying Xue
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Zhi-Yu Zhu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yue Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
16
|
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
|
17
|
Tsubokawa D. Immunomodulators secreted from parasitic helminths act on pattern recognition receptors. FRONTIERS IN PARASITOLOGY 2023; 1:1091596. [PMID: 39816467 PMCID: PMC11731691 DOI: 10.3389/fpara.2022.1091596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2025]
Abstract
Excretory-secretory (ES) products from parasitic helminths contain immunomodulatory molecules, which can regulate host immune responses. These immunomodulatory molecules are crucial for successful parasitism, and play roles in tissue migration, maturation, and reproduction. Some target pattern recognition receptors (PRRs), including toll-like receptor, C-type lectin receptor, receptor for advanced glycation end products, and nucleotide-binding oligomerization domain-like receptor. PRRs trigger activation of signaling cascades, inducing innate inflammatory responses and adaptive immunity in hosts. This article reviews ES immunomodulators identified in parasitic helminths that act on PRRs, and their PRR-facilitated immune-regulatory mechanisms. In addition, we describe the therapeutic potential of ES immunomodulators for allergic and inflammatory diseases.
Collapse
Affiliation(s)
- Daigo Tsubokawa
- Department of Parasitology and Tropical Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
18
|
Chenuet P, Marquant Q, Fauconnier L, Youness A, Mellier M, Marchiol T, Rouxel N, Messaoud-Nacer Y, Maillet I, Ledru A, Quesniaux VFJ, Ryffel B, Horsnell W, Végran F, Apetoh L, Togbe D. NLRP6 negatively regulates type 2 immune responses in mice. Allergy 2022; 77:3320-3336. [PMID: 35615773 DOI: 10.1111/all.15388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/05/2022] [Accepted: 05/02/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Inflammasomes are large protein complexes that assemble in the cytosol in response to danger such as tissue damage or infection. Following activation, inflammasomes trigger cell death and the release of biologically active forms of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. NOD-like receptor family pyrin domain containing 6 (NLRP6) inflammasome is required for IL-18 secretion by intestinal epithelial cells, macrophages, and T cells, contributing to homeostasis and self-defense against pathogenic microbes. However, the involvement of NLRP6 in type 2 lung inflammation remains elusive. METHODS Wild-type (WT) and Nlrp6-/- mice were used. Birch pollen extract (BPE)-induced allergic lung inflammation, eosinophil recruitment, Th2-related cytokine and chemokine production, airway hyperresponsiveness, and lung histopathology, Th2 cell differentiation, GATA3, and Th2 cytokines expression, were determined. Nippostrongylus brasiliensis (Nb) infection, worm count in intestine, type 2 innate lymphoid cell (ILC2), and Th2 cells in lungs were evaluated. RESULTS We demonstrate in Nlrp6-/- mice that a mixed Th2/Th17 immune responses prevailed following birch pollen challenge with increased eosinophils, ILC2, Th2, and Th17 cell induction and reduced IL-18 production. Nippostrongylus brasiliensis infected Nlrp6-/- mice featured enhanced early expulsion of the parasite due to enhanced type 2 immune responses compared to WT hosts. In vitro, NLRP6 repressed Th2 polarization, as shown by increased Th2 cytokines and higher expression of the transcription factor GATA3 in the absence of NLRP6. Exogenous IL-18 administration partially reduced the enhanced airways inflammation in Nlrp6-/- mice. CONCLUSIONS In summary, our data identify NLRP6 as a negative regulator of type 2 immune responses.
Collapse
Affiliation(s)
| | - Quentin Marquant
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| | | | - Ali Youness
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| | | | | | | | - Yasmine Messaoud-Nacer
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| | - Isabelle Maillet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| | | | - Valérie F J Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| | - William Horsnell
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France.,Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town 7925, South Africa & South African Medical Research Council, Cape Town, South Africa.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | | | - Dieudonnée Togbe
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans-Cedex 2, France
| |
Collapse
|
19
|
Abuzeid AMI, Hefni MM, Huang Y, He L, Zhuang T, Li G. Immune pathogenesis in pigeons during experimental Prohemistomum vivax infection. Front Vet Sci 2022; 9:974698. [PMID: 36187827 PMCID: PMC9516004 DOI: 10.3389/fvets.2022.974698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Prohemistomum vivax is a small trematode belonging to the family Cyathocotylidae, infecting fish-eating birds and mammals, including humans. However, no data on molecular identification and immune pathogenesis are available, challenging effective diagnostic and therapeutic interventions. Here, we identified P. vivax based on combined morphological and molecular data and examined histopathological lesions and the differential cytokines expression in experimentally infected pigeons. Pigeons were orally infected with 500 prohemistomid metacercariae. Intestinal and spleen tissues were harvested 2, 4, 7, 14, 21, and 28 days post-infection (dpi). Gene expression levels of eleven cytokines (IL-1, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, IL-15, IL-18, IFN-γ, and TGF-β3) were assessed using quantitative reverse-transcription PCR (RT-qPCR). We identified the recovered flukes as Prohemistomum vivax based on morphological features and the sequence and phylogenetic analysis of the internal transcribed spacer 1 (ITS1), 5.8 ribosomal RNA, and ITS2 region. Histopathological lesions were induced as early as 2 dpi, with the intensity of villi atrophy and inflammatory cell infiltration increasing as the infection progressed. An early immunosuppressive state (2 and 4 dpi), with TGF-β3 overexpression, developed to allow parasite colonization. A mixed Th1/Th2 immune response (overexpressed IFN-γ, IL-12, IL-2, IL-4, and IL-5) was activated as the infection progressed from 7 to 28 dpi. Inflammatory cytokines (IL-1, IL-6, IL-18, and IL-15) were generally overexpressed at 7–28 dpi, peaking at 7 or 14 dpi. The upregulated Treg IL-10 expression peaking between 21 and 28 dpi might promote the Th1/Th2 balance and immune homeostasis to protect the host from excessive tissue pathology and inflammation. The intestine and spleen expressed a significantly different relative quantity of cytokines throughout the infection. To conclude, our results presented distinct cytokine alteration throughout P. vivax infection in pigeons, which may aid in understanding the immune pathogenesis and host defense mechanism against this infection.
Collapse
Affiliation(s)
- Asmaa M. I. Abuzeid
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Department of Parasitology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Mahmoud M. Hefni
- Institute of Biotechnology for Postgraduates Studies and Researches, Suez Canal University, Ismailia, Egypt
- Mahmoud M. Hefni
| | - Yue Huang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Long He
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Tingting Zhuang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guoqing Li
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- *Correspondence: Guoqing Li
| |
Collapse
|
20
|
Sylvester M, Son A, Schwartz DM. The Interactions Between Autoinflammation and Type 2 Immunity: From Mechanistic Studies to Epidemiologic Associations. Front Immunol 2022; 13:818039. [PMID: 35281022 PMCID: PMC8907424 DOI: 10.3389/fimmu.2022.818039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/02/2022] [Indexed: 12/30/2022] Open
Abstract
Autoinflammatory diseases are a group of clinical syndromes characterized by constitutive overactivation of innate immune pathways. This results in increased production of or responses to monocyte- and neutrophil-derived cytokines such as interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), and Type 1 interferon (IFN). By contrast, clinical allergy is caused by dysregulated type 2 immunity, which is characterized by expansion of T helper 2 (Th2) cells and eosinophils, as well as overproduction of the associated cytokines IL-4, IL-5, IL-9, and IL-13. Traditionally, type 2 immune cells and autoinflammatory effectors were thought to counter-regulate each other. However, an expanding body of evidence suggests that, in some contexts, autoinflammatory pathways and cytokines may potentiate type 2 immune responses. Conversely, type 2 immune cells and cytokines can regulate autoinflammatory responses in complex and context-dependent manners. Here, we introduce the concepts of autoinflammation and type 2 immunity. We proceed to review the mechanisms by which autoinflammatory and type 2 immune responses can modulate each other. Finally, we discuss the epidemiology of type 2 immunity and clinical allergy in several monogenic and complex autoinflammatory diseases. In the future, these interactions between type 2 immunity and autoinflammation may help to expand the spectrum of autoinflammation and to guide the management of patients with various autoinflammatory and allergic diseases.
Collapse
Affiliation(s)
- McKella Sylvester
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aran Son
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
21
|
Sanches RCO, Mambelli F, Oliveira SC. Neutrophils and schistosomiasis: a missing piece in pathology. Parasite Immunol 2022; 44:e12916. [PMID: 35332932 DOI: 10.1111/pim.12916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022]
Abstract
Schistosomiasis is a chronic human parasitic disease that causes serious health problems worldwide. The disease-associated liver pathology is one of the hallmarks of infections by S. mansoni and S. japonicum, and is accountable for the debilitating condition found in infected patients. In the past few years, investigative studies have highlighted the key role played by neutrophils and the influence of inflammasome signaling pathway in different pathological conditions. However, it is noteworthy that the study of inflammasome activation in neutrophils has been overlooked by reports concerning macrophages and monocytes. This interplay between neutrophils and inflammasomes is much more poorly investigated during schistosomiasis. Herein we reviewed the role of neutrophils during schistosomiasis and addressed the potential connection between these cells and inflammasome activation in this context.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio Mambelli
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, Salvador, Brazil
| |
Collapse
|
22
|
Martynova E, Rizvanov A, Urbanowicz RA, Khaiboullina S. Inflammasome Contribution to the Activation of Th1, Th2, and Th17 Immune Responses. Front Microbiol 2022; 13:851835. [PMID: 35369454 PMCID: PMC8969514 DOI: 10.3389/fmicb.2022.851835] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are cytosolic polyprotein complexes formed in response to various external and internal stimuli, including viral and bacterial antigens. The main product of the inflammasome is active caspase 1 which proteolytically cleaves, releasing functional interleukin-1 beta (IL-1β) and interleukin-18 (IL-18). These cytokines play a central role in shaping immune response to pathogens. In this review, we will focus on the mechanisms of inflammasome activation, as well as their role in development of Th1, Th2, and Th17 lymphocytes. The contribution of cytokines IL-1β, IL-18, and IL-33, products of activated inflammasomes, are summarized. Additionally, the role of cytokines released from tissue cells in promoting differentiation of lymphocyte populations is discussed.
Collapse
Affiliation(s)
| | | | - Richard A. Urbanowicz
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
23
|
Yang Q, Ouyang J, Pi D, Feng L, Yang J. Malassezia in Inflammatory Bowel Disease: Accomplice of Evoking Tumorigenesis. Front Immunol 2022; 13:846469. [PMID: 35309351 PMCID: PMC8931276 DOI: 10.3389/fimmu.2022.846469] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/10/2022] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence indicates that patients with inflammatory bowel disease (IBD) have a significantly higher risk of developing different cancers, while the exact mechanism involved is not yet fully understood. Malassezia is a lipid-dependent opportunistic yeast, which colonizes on mammalian skin and internal organs. Also, dysbiosis in fungal communities accompanied by high level of Malassezia are fairly common in inflammatory diseases such as IBD and various cancers. In cancer patients, higher levels of Malassezia are associated with worse prognosis. Once it is ablated in tumor-bearing mice, their prognostic conditions will be improved. Moreover, Malassezia manifests multiple proinflammatory biological properties, such as destruction of epithelial barrier, enrichment of inflammatory factors, and degradation of extracellular matrix (ECM), all of which have been reported to contribute to tumor initiation and malignant progression. Based on these facts, we hypothesize that high levels of Malassezia together with mycobiome dysbiosis in patients with IBD, would aggravate the microecological imbalance, worsen the inflammatory response, and further promote tumorigenesis and deterioration. Herein, we will discuss the detrimental properties of Malassezia and explore the key role of this fungus in the correlation between IBD and cancer, in order to take early surveillance and intervention to minimize the cancer risk in individuals with IBD.
Collapse
Affiliation(s)
- Qiyu Yang
- Department of Radiation Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Jing Ouyang
- Chongqing Public Health Medical Center, Chongqing, China
| | - Damao Pi
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Feng
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
- *Correspondence: Li Feng, ; Jiadan Yang,
| | - Jiadan Yang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Li Feng, ; Jiadan Yang,
| |
Collapse
|
24
|
Li Z, Chen X, Tao J, Shi A, Zhang J, Yu P. Exosomes Regulate NLRP3 Inflammasome in Diseases. Front Cell Dev Biol 2022; 9:802509. [PMID: 35047512 PMCID: PMC8762245 DOI: 10.3389/fcell.2021.802509] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence has suggested the unique and critical role of exosomes as signal molecules vector in various diseases. Numerous researchers have been trying to identify how these exosomes function in immune progression, as this could promote their use as biomarkers for the disease process and potential promising diagnostic tools. NOD-like receptor (NLR) family, pyrin domain containing 3 (NLRP3), a tripartite protein, contains three functional domains a central nucleotide-binding and oligomerization domain (NACHT), an N-terminal pyrin domain (PYD), and a leucine-rich repeat domain (LRR). Of note, existing studies have identified exosome as a novel mediator of the NLRP3 inflammasome, which is critical in diseases progression. However, the actual mechanisms and clinical treatment related to exosomes and NLRP3 are still not fully understood. Herein, we presented an up-to-date review of exosomes and NLRP3 in diseases, outlining what is known about the role of exosomes in the activation of NLRP3 inflammasome and also highlighting areas of this topic that warrant further study.
Collapse
Affiliation(s)
- Zhangwang Li
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Xinyue Chen
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Junjie Tao
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Ao Shi
- School of Medicine, University of Nicosia, Nicosia, Cyprus.,School of Medicine, St. George University of London, London, United Kingdom
| | - Jing Zhang
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China.,Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China.,Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Age-dependent rise in IFN-γ competence undermines effective type 2 responses to nematode infection. Mucosal Immunol 2022; 15:1270-1282. [PMID: 35690651 PMCID: PMC9705248 DOI: 10.1038/s41385-022-00519-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023]
Abstract
The efficient induction of type 2 immune responses is central to the control of helminth infections. Previous studies demonstrated that strong Th1 responses driven by intracellular pathogens as well as a bias for type 1 activity in senescent mice impedes the generation of Th2 responses and the control of intestinal nematode infections. Here, we show that the spontaneous differentiation of Th1 cells and their expansion with age restrains type 2 immunity to infection with the small intestinal nematode H. polygyrus much earlier in life than previously anticipated. This includes the more extensive induction of IFN-γ competent, nematode-specific Th2/1 hybrid cells in BALB/c mice older than three months compared to younger animals. In C57BL/6 mice, Th1 cells accumulate more rapidly at steady state, translating to elevated Th2/1 differentiation and poor control of parasite fitness in primary infections experienced at a young age. Blocking of early IFN-γ and IL-12 signals during the first week of nematode infection leads to sharply decreased Th2/1 differentiation and promotes resistance in both mouse lines. Together, these data suggest that IFN-γ competent, type 1 like effector cells spontaneously accumulating in the vertebrate host progressively curtail the effectiveness of anti-nematode type 2 responses with rising host age.
Collapse
|
26
|
Babamale AO, Chen ST. Nod-like Receptors: Critical Intracellular Sensors for Host Protection and Cell Death in Microbial and Parasitic Infections. Int J Mol Sci 2021; 22:11398. [PMID: 34768828 PMCID: PMC8584118 DOI: 10.3390/ijms222111398] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/04/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Cell death is an essential immunological apparatus of host defense, but dysregulation of mutually inclusive cell deaths poses severe threats during microbial and parasitic infections leading to deleterious consequences in the pathological progression of infectious diseases. Nucleotide-binding oligomerization domain (NOD)-Leucine-rich repeats (LRR)-containing receptors (NLRs), also called nucleotide-binding oligomerization (NOD)-like receptors (NLRs), are major cytosolic pattern recognition receptors (PRRs), their involvement in the orchestration of innate immunity and host defense against bacteria, viruses, fungi and parasites, often results in the cleavage of gasdermin and the release of IL-1β and IL-18, should be tightly regulated. NLRs are functionally diverse and tissue-specific PRRs expressed by both immune and non-immune cells. Beyond the inflammasome activation, NLRs are also involved in NF-κB and MAPK activation signaling, the regulation of type I IFN (IFN-I) production and the inflammatory cell death during microbial infections. Recent advancements of NLRs biology revealed its possible interplay with pyroptotic cell death and inflammatory mediators, such as caspase 1, caspase 11, IFN-I and GSDMD. This review provides the most updated information that caspase 8 skews the NLRP3 inflammasome activation in PANoptosis during pathogen infection. We also update multidimensional roles of NLRP12 in regulating innate immunity in a content-dependent manner: novel interference of NLRP12 on TLRs and NOD derived-signaling cascade, and the recently unveiled regulatory property of NLRP12 in production of type I IFN. Future prospects of exploring NLRs in controlling cell death during parasitic and microbial infection were highlighted.
Collapse
Affiliation(s)
- Abdulkareem Olarewaju Babamale
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming Chiao Tung University and Academia Sinica, Taipei 11266, Taiwan;
- Parasitology Unit, Faculty of Life Sciences, University of Ilorin, Ilorin 240003, Nigeria
| | - Szu-Ting Chen
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming Chiao Tung University and Academia Sinica, Taipei 11266, Taiwan;
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 11266, Taiwan
- Cancer Progression Research Center, National Yang-Ming Chiao Tung University, Taipei 11266, Taiwan
| |
Collapse
|
27
|
Corcoran SE, Halai R, Cooper MA. Pharmacological Inhibition of the Nod-Like Receptor Family Pyrin Domain Containing 3 Inflammasome with MCC950. Pharmacol Rev 2021; 73:968-1000. [PMID: 34117094 DOI: 10.1124/pharmrev.120.000171] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Activation of the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome drives release of the proinflammatory cytokines interleukin (IL)-1β and IL-18 and induces pyroptosis (lytic cell death). These events drive chronic inflammation, and as such, NLRP3 has been implicated in a large number of human diseases. These range from autoimmune conditions, the simplest of which is NLRP3 gain-of-function mutations leading to an orphan disease, cryopyrin-associated period syndrome, to large disease burden indications, such as atherosclerosis, heart failure, stroke, neurodegeneration, asthma, ulcerative colitis, and arthritis. The potential clinical utility of NLRP3 inhibitors is substantiated by an expanding list of indications in which NLRP3 activation has been shown to play a detrimental role. Studies of pharmacological inhibition of NLRP3 in nonclinical models of disease using MCC950 in combination with human genetics, epigenetics, and analyses of the efficacy of biologic inhibitors of IL-1β, such as anakinra and canakinumab, can help to prioritize clinical trials of NLRP3-directed therapeutics. Although MCC950 shows excellent (nanomolar) potency and high target selectivity, its pharmacokinetic and toxicokinetic properties limited its therapeutic development in the clinic. Several improved, next-generation inhibitors are now in clinical trials. Hence the body of research in a plethora of conditions reviewed herein may inform analysis of the potential translational value of NLRP3 inhibition in diseases with significant unmet medical need. SIGNIFICANCE STATEMENT: The nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is one of the most widely studied and best validated biological targets in innate immunity. Activation of NLRP3 can be inhibited with MCC950, resulting in efficacy in more than 100 nonclinical models of inflammatory diseases. As several next-generation NLRP3 inhibitors are entering proof-of-concept clinical trials in 2020, a review of the pharmacology of MCC950 is timely and significant.
Collapse
Affiliation(s)
- Sarah E Corcoran
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Reena Halai
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Matthew A Cooper
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| |
Collapse
|
28
|
Huanosta-Murillo E, Alcántara-Hernández M, Hernández-Rico B, Victoria-Acosta G, Miranda-Cruz P, Domínguez-Gómez MA, Jurado-Santacruz F, Patiño-López G, Pérez-Koldenkova V, Palma-Guzmán A, Licona-Limón P, Fuentes-Pananá EM, Lemini-López A, Bonifaz LC. NLRP3 Regulates IL-4 Expression in TOX + CD4 + T Cells of Cutaneous T Cell Lymphoma to Potentially Promote Disease Progression. Front Immunol 2021; 12:668369. [PMID: 34220814 PMCID: PMC8244903 DOI: 10.3389/fimmu.2021.668369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/31/2021] [Indexed: 11/27/2022] Open
Abstract
In cutaneous T cell lymphoma (CTCL), a dominant Th2 profile associated with disease progression has been proposed. Moreover, although the production and regulation of IL-4 expression during the early stages of the disease may have important implications in later stages, these processes are poorly understood. Here, we demonstrate the presence of TOX+ CD4+ T cells that produce IL-4+ in early-stage skin lesions of CTCL patients and reveal a complex mechanism by which the NLRP3 receptor promotes a Th2 response by controlling IL-4 production. Unassembled NLRP3 is able to translocate to the nucleus of malignant CD4+ T cells, where it binds to the human il-4 promoter. Accordingly, IL-4 expression is decreased by knocking down and increased by promoting the nuclear localization of NLRP3. We describe a positive feedback loop in which IL-4 inhibits NLRP3 inflammasome assembly, thereby further increasing its production. IL-4 induced a potentially malignant phenotype measured based on TOX expression and proliferation. This mechanism of IL-4 regulation mediated by NLRP3 is amplified in late-stage CTCL associated with disease progression. These results indicate that NLRP3 might be a key regulator of IL-4 expression in TOX+ CD4+ T cells of CTCL patients and that this mechanism might have important implications in the progression of the disease.
Collapse
MESH Headings
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cytotoxicity, Immunologic
- Disease Progression
- Gene Expression Regulation, Neoplastic
- Humans
- Interleukin-4/genetics
- Interleukin-4/metabolism
- Jurkat Cells
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/immunology
- Lymphoma, T-Cell, Cutaneous/metabolism
- Mexico
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Phenotype
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
Collapse
Affiliation(s)
- Enrique Huanosta-Murillo
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Marcela Alcántara-Hernández
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| | - Brenda Hernández-Rico
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Patricia Miranda-Cruz
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Fermín Jurado-Santacruz
- Centro Dermatológico Dr. Ladislao de la Pascua, Secretaría de Salud de la Ciudad de México, Mexico City, Mexico
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Sección de Biología Celular de Linfocitos, Unidad de Hemato-Oncología e Investigación Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Vadim Pérez-Koldenkova
- Laboratorio Nacional de Microscopía Avanzada, División de Desarrollo de la Investigación, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Alam Palma-Guzmán
- Laboratorio de Histología, Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ezequiel M. Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Alicia Lemini-López
- Servicio de Dermatología, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
29
|
Abstract
A fundamental concept in immunology is that the innate immune system initiates or instructs downstream adaptive immune responses. Inflammasomes are central players in innate immunity to pathogens, but how inflammasomes shape adaptive immunity is complex and relatively poorly understood. Here we highlight recent work on the interplay between inflammasomes and adaptive immunity. We address how inflammasome-dependent release of cytokines and antigen activates, shapes or even inhibits adaptive immune responses. We consider how distinct tissue or cellular contexts may alter the effects of inflammasome activation on adaptive immunity and how this contributes to beneficial or detrimental outcomes in infectious diseases, cancer and autoimmunity. We aspire to provide a framework for thinking about inflammasomes and their connection to the adaptive immune response.
Collapse
|
30
|
Darlan DM, Rozi MF, Yulfi H. Overview of Immunological Responses and Immunomodulation Properties of Trichuris sp.: Prospects for Better Understanding Human Trichuriasis. Life (Basel) 2021; 11:188. [PMID: 33673676 PMCID: PMC7997218 DOI: 10.3390/life11030188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 12/23/2022] Open
Abstract
Trichuris sp. infection has appeared as a pathological burden in the population, but the immunomodulation features could result in an opportunity to discover novel treatments for diseases with prominent inflammatory responses. Regarding the immunological aspects, the innate immune responses against Trichuris sp. are also responsible for determining subsequent immune responses, including the activation of innate lymphoid cell type 2 (ILC2s), and encouraging the immune cell polarization of the resistant host phenotype. Nevertheless, this parasite can establish a supportive niche for worm survival and finally avoid host immune interference. Trichuris sp. could skew antigen recognition and immune cell activation and proliferation through the generation of specific substances, called excretory/secretory (ESPs) and soluble products (SPs), which mainly mediate its immunomodulation properties. Through this review, we elaborate and discuss innate-adaptive immune responses and immunomodulation aspects, as well as the clinical implications for managing inflammatory-based diseases, such as inflammatory bowel diseases, allergic, sepsis, and other autoimmune diseases.
Collapse
Affiliation(s)
- Dewi Masyithah Darlan
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.D.); (H.Y.)
| | | | - Hemma Yulfi
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.D.); (H.Y.)
| |
Collapse
|
31
|
Soare AY, Freeman TL, Min AK, Malik HS, Osota EO, Swartz TH. P2RX7 at the Host-Pathogen Interface of Infectious Diseases. Microbiol Mol Biol Rev 2021; 85:e00055-20. [PMID: 33441488 PMCID: PMC7849353 DOI: 10.1128/mmbr.00055-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The P2X7 receptor (P2RX7) is an important molecule that functions as a danger sensor, detecting extracellular nucleotides from injured cells and thus signaling an inflammatory program to nearby cells. It is expressed in immune cells and plays important roles in pathogen surveillance and cell-mediated responses to infectious organisms. There is an abundance of literature on the role of P2RX7 in inflammatory diseases and the role of these receptors in host-pathogen interactions. Here, we describe the current knowledge of the role of P2RX7 in the host response to a variety of pathogens, including viruses, bacteria, fungi, protozoa, and helminths. We describe in vitro and in vivo evidence for the critical role these receptors play in mediating and modulating immune responses. Our observations indicate a role for P2X7 signaling in sensing damage-associated molecular patterns released by nearby infected cells to facilitate immunopathology or protection. In this review, we describe how P2RX7 signaling can play critical roles in numerous cells types in response to a diverse array of pathogens in mediating pathogenesis and immunity to infectious agents.
Collapse
Affiliation(s)
- Alexandra Y Soare
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tracey L Freeman
- Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alice K Min
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hagerah S Malik
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Elizabeth O Osota
- University of California San Diego, Graduate School of Biomedical Sciences, San Diego, California, USA
| | - Talia H Swartz
- Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
32
|
Zhang LJ, Wang XD, Ji MS, Hasimu H, Abliz P. Characterisation of a clinical isolated Aspergillus lentulus strain using a Galleria mellonella infection model. J Thorac Dis 2021; 13:803-811. [PMID: 33717553 PMCID: PMC7947491 DOI: 10.21037/jtd-20-961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/03/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND In recent years, the number of invasive aspergillus infection cases caused by Aspergillus lentulus (A. lentulus) has gradually increased and this fungus is usually difficult to distinguish from Aspergillus fumigatus in morphology. All of these presents a great challenge to the treatment of invasive fungal infections caused by A. lentulus. The present study aims to discuss the antifungal resistance, virulence and inflammatory factors' changes after the infection of larvae of A. lentulus separated from patients with chronic obstructive pulmonary disease (COPD) to reflect the host immune response. METHODS A. lentulus isolated from COPD patients was identified by morphology and molecular biology, and its drug sensitivity was determined in vitro. Then the virulence factors and inflammatory response related factors of A. lentulus were determined by the model of A. lentulus infecting larvae. These were divided into three groups: A. lentulus standard strain, A. lentulus strain isolated from patients; PBS control. The infection model was formed by injecting the suspension of A. lentulus at a concentration of 1×106 CFU into larvae, in order to determine the (1,3)-β-D-glucan and galactomannan levels, and determine the caspase-1 and TNF-α concentration in Galleria mellonella larvae by RT-PCR. RESULTS The results revealed that A. lentulus had good sensitivity to itraconazole, voriconazole and micafungin, while (1,3)-β-D-glucan was negative in the two groups. The level of galactomannan in the two groups was higher than that in the control group, and the difference was statistically significant (P<0.05). However, there was no statistical difference between the standard strain group and patient strain group (P>0.05). After the infection of larvae, caspase-1 and TNF-α in the Galleria mellonella larvae increased in the two groups, and these elevated levels were statistically significant in both groups (P<0.05). However, there was no significant difference between the two groups (P>0.05). CONCLUSIONS There is no significant difference in virulence factor and host inflammatory response between A. lentulus isolated from COPD patients and standard strains. Galactomannan has more advantages in the early detection of A. lentulus invasive infection. Furthermore, the caspase-1-mediated inflammasome pathway may be involved in the host immune response to A. lentulus.
Collapse
Affiliation(s)
- Li-Juan Zhang
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiao-Dong Wang
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ming-Shuo Ji
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hadiliya Hasimu
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Paride Abliz
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
33
|
Park HR, Oh JH, Lee YJ, Park SH, Lee YW, Lee S, Kang H, Kim JE. Inflammasome-mediated Inflammation by Malassezia in human keratinocytes: A comparative analysis with different strains. Mycoses 2020; 64:292-299. [PMID: 33206994 DOI: 10.1111/myc.13214] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 01/03/2023]
Abstract
Malassezia species are associated with several common dermatologic conditions including pityriasis versicolor, seborrhoeic dermatitis, folliculitis, and atopic dermatitis and dandruff. However, its causal role remains to be established. We intended to explore the role of inflammasome activation in human keratinocytes in response to three different Malassezia species. We compared the different activation patterns of inflammasomes and the expression of pro-inflammatory cytokines and antimicrobial peptides by three different Malassezia species-M. restricta, M. globosa and M. sympodialis-in human keratinocytes. We found that different Malassezia species, especially M. restricta and M. globosa could induce nucleotide-binding oligomerisation domain, leucine-rich repeat and pyrin-domain-containing protein (NLRP)3-apoptosis-associated speck-like protein containing CARD (ASC) inflammasome activation and subsequent interleukin (IL)-1β secretion in human keratinocytes. Malassezia species variably induced thymic stromal lymphopoietin, β-defensin 2, and LL-37. IL-8 mRNA and IL-22 protein significantly increased in the M. sympodialis-treated group, and Chemokine C-C motif ligand (CCL)17 and CCL22 mRNA were increased in response to M. globosa- and M. restricta- treated keratinocytes, respectively. Our data show that various species of Malassezia promote variable inflammatory responses in keratinocytes by activating NLRP3 inflammasomes, pro-inflammatory cytokines and chemokines, and antimicrobial peptides.
Collapse
Affiliation(s)
- Hye Ree Park
- Department of Dermatology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jee Hye Oh
- Department of Dermatology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Yu Jin Lee
- Department of Dermatology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Song Hee Park
- Department of Dermatology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Yang Won Lee
- Department of Dermatology, Konkuk University School of Medicine, Seoul, Korea.,Research Institute of Medical Science, Konkuk University, Seoul, Korea
| | - Seongju Lee
- Department of Anatomy and Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon, South Korea
| | - Hoon Kang
- Department of Dermatology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jung Eun Kim
- Department of Dermatology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
34
|
Heterogeneity in the initiation, development and function of type 2 immunity. Nat Rev Immunol 2020; 20:603-614. [PMID: 32367051 PMCID: PMC9773851 DOI: 10.1038/s41577-020-0301-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Type 2 immune responses operate under varying conditions in distinct tissue environments and are crucial for protection against helminth infections and for the maintenance of tissue homeostasis. Here we explore how different layers of heterogeneity influence type 2 immunity. Distinct insults, such as allergens or infections, can induce type 2 immune responses through diverse mechanisms, and this can have heterogeneous consequences, ranging from acute or chronic inflammation to deficits in immune regulation and tissue repair. Technological advances have provided new insights into the molecular heterogeneity of different developmental lineages of type 2 immune cells. Genetic and environmental heterogeneity also contributes to the varying magnitude and quality of the type 2 immune response during infection, which is an important determinant of the balance between pathology and disease resolution. Hence, understanding the mechanisms underlying the heterogeneity of type 2 immune responses between individuals and between different tissues will be crucial for treating diseases in which type 2 immunity is an important component.
Collapse
|
35
|
Activation of the NLRP3 Inflammasome by Particles from the Echinococcus granulosus Laminated Layer. Infect Immun 2020; 88:IAI.00190-20. [PMID: 32571988 PMCID: PMC7440765 DOI: 10.1128/iai.00190-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022] Open
Abstract
The interaction of dendritic cells and macrophages with a variety of rigid noncellular particles triggers activation of the NLRP3 inflammasome and consequent secretion of interleukin 1β (IL-1β). Noncellular particles can also be generated in the context of helminth infection, since these large pathogens often shed their outermost structures during growth and/or molting. One such structure is the massive, mucin-based, soft, flexible laminated layer (LL), which protects the larval stages of cestodes of the genus Echinococcus. The interaction of dendritic cells and macrophages with a variety of rigid noncellular particles triggers activation of the NLRP3 inflammasome and consequent secretion of interleukin 1β (IL-1β). Noncellular particles can also be generated in the context of helminth infection, since these large pathogens often shed their outermost structures during growth and/or molting. One such structure is the massive, mucin-based, soft, flexible laminated layer (LL), which protects the larval stages of cestodes of the genus Echinococcus. We show that particles from the Echinococcus granulosus LL (pLL) trigger NLRP3- and caspase-1-dependent IL-1β in lipopolysaccharide (LPS)-primed mouse bone marrow-derived dendritic cells (BMDC). This response can be elicited by pLL too large for phagocytosis and nonetheless requires actin dynamics, Syk, and phosphatidylinositol 3-kinase (PI3K). These three requirements had already been observed in our previous study on the alteration by pLL of CD86, CD40, IL-10, and IL-12 responses to LPS in BMDC; however, we now show that these alterations are independent of NLRP3 and caspase-1. In other words, an initial interaction with particles requiring actin dynamics, Syk, and PI3K, but not phagocytosis, elicits both NLRP3-dependent and NLRP3-independent responses. Intraperitoneal injection of pLL induced IL-1β, suggesting that contact with LL materials induces IL-1β in the E. granulosus infection setting. Our results extend our understanding of NLRP3 inflammasome activation by noncellular particulate materials both to helminth-derived materials and to flexible/soft materials.
Collapse
|
36
|
Iacob SA, Olariu MC, Iacob DG. Eosinophilic Colitis and Clostridioides difficile Sepsis With Rapid Remission After Antimicrobial Treatment; A Rare Coincidence and Its Pathogenic Implications. Front Med (Lausanne) 2020; 7:328. [PMID: 32903297 PMCID: PMC7396602 DOI: 10.3389/fmed.2020.00328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/03/2020] [Indexed: 11/24/2022] Open
Abstract
Eosinophilic colitis is a rare inflammatory disorder of the digestive tract with chronic evolution and unknown pathophysiological mechanisms. The article describes the case of a 64-year old woman with a history of asthma and hypereosinophilia, who presented to a surgical department for persistent abdominal pain in the past 4 months, weight loss and malabsorption. She was diagnosed with eosinophilic colitis based on the colonoscopic result indicating extensive eosinophilic infiltration of the colonic mucosa correlated with the laboratory data and abdominal CT scan results. Following the colonoscopy, the patient developed fever, hypotension and diarrhea and was transferred to an Infectious Diseases Department with a presumptive diagnosis of abdominal sepsis. Treatment with ertapenem was immediately started. Metronidazole was also added due to a PCR positive stool test for Clostridioides difficile toxins encoding-genes. The patient displayed a rapid remission of the fever and of the intestinal complaints following antibiotic therapy and was discharged after 14 days. During a 3 months follow-up, the patient remained asymptomatic with normal values of laboratory parameters except for a persistent hypereosinophilia. The case outlines two distinguishing features: a histopathologic diagnosis of eosinophilic colitis, a rare diagnosis of a patient with chronic abdominal pain and an unexpected and rapid remission of the eosinophilic colitis following the antibiotic treatment and the restoration of the intestinal eubiosis.
Collapse
Affiliation(s)
- Simona Alexandra Iacob
- Infectious Diseases Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Infectious Diseases Department, The National Institute of Infectious Diseases "Matei Bals", Bucharest, Romania
| | - Mihaela Cristina Olariu
- Infectious Diseases Department, The National Institute of Infectious Diseases "Matei Bals", Bucharest, Romania.,Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
37
|
Bakhtiar NM, Spotin A, Mahami-Oskouei M, Ahmadpour E, Rostami A. Recent advances on innate immune pathways related to host-parasite cross-talk in cystic and alveolar echinococcosis. Parasit Vectors 2020; 13:232. [PMID: 32375891 PMCID: PMC7204293 DOI: 10.1186/s13071-020-04103-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic echinococcosis (CE) and alveolar echinococcosis (AE) are life-threatening parasitic infections worldwide caused by Echinococcus granulosus (sensu lato) and E. multilocularis, respectively. Very little is known about the factors affecting innate susceptibility and resistance to infection with Echinococcus spp. Although benzimidazolic drugs against CE and AE have definitively improved the treatment of these cestodes; however, the lack of successful control campaigns, including the EG95 vaccine, at a continental level indicates the importance of generating novel therapies. This review represents an update on the latest developments in the regulatory functions of innate immune pathways such as apoptosis, toll-like receptors (TLRs), and inflammasomes against CE and AE. We suggest that apoptosis can reciprocally play a bi-functional role among the host-Echinococcus metabolite relationships in suppressive and survival mechanisms of CE. Based on the available information, further studies are needed to determine whether the orchestrated in silico strategy for designing inhibitors and interfering RNA against anti-apoptotic proteins and TLRs would be effective to improve new treatments as well as therapeutic vaccines against the E. granulosus and E. multilocularis.![]()
Collapse
Affiliation(s)
- Nayer Mehdizad Bakhtiar
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Spotin
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahmoud Mahami-Oskouei
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Ahmadpour
- Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
38
|
Abstract
Fasciola hepatica, the common liver fluke, causes infection of livestock throughout temperate regions of the globe. This helminth parasite has an indirect lifecycle, relying on the presence of the mud snail to complete its transition from egg to definitive host (Beesley et al., Transbound Emerg Dis 65:199-216, 2017). Within the definitive host, the parasite excysts in the intestine forming a newly excysted juvenile (NEJ) and migrates via the peritoneal cavity to the liver. Disease resulting from infection can be acute or chronic depending on the host and the number of parasites present. Sheep may succumb to a fatal acute infection if the challenge of metacercariae is great enough. However, in cattle chronic disease is the most likely outcome with parasites surviving for long periods of time. Annual losses are estimated to be in the region of US$ 2000 million to the agricultural industry (Beesley et al., Transbound Emerg Dis 65:199-216, 2017). Management of the disease depends heavily on chemotherapy with triclabendazole being the drug of choice, consistent use for over 20 years has resulted in drug-resistant strains emerging worldwide (Beesley et al., Int J Parasitol 47:11-20, 2017). A more sustainable approach to control would be through vaccination and indeed a lead candidate has been identified, cathepsin L1. Despite these promising results the parasite continues to confound our own and host efforts to generate long-lasting and effective immunity. In this brief review we focus our attention on those mechanisms that the parasite utilises to circumvent the innate based defense mechanisms within the host.
Collapse
Affiliation(s)
- Robin J Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.
| | - Mayowa Musah-Eroje
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| |
Collapse
|
39
|
Celias DP, Motrán CC, Cervi L. Helminths Turning on the NLRP3 Inflammasome: Pros and Cons. Trends Parasitol 2019; 36:87-90. [PMID: 31753545 DOI: 10.1016/j.pt.2019.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 01/24/2023]
Abstract
Helminth infections can activate multimeric protein complexes called inflammasomes. In this forum we summarize the main effects of the NLRP3 inflammasome activation, including control of excessive Th2 response and immunopathology induction. These mechanisms would ensure the survival of both the host and the parasite.
Collapse
Affiliation(s)
- Daiana Pamela Celias
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina.
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina.
| |
Collapse
|
40
|
Chenery AL, Alhallaf R, Agha Z, Ajendra J, Parkinson JE, Cooper MM, Chan BHK, Eichenberger RM, Dent LA, Robertson AAB, Kupz A, Brough D, Loukas A, Sutherland TE, Allen JE, Giacomin PR. Inflammasome-Independent Role for NLRP3 in Controlling Innate Antihelminth Immunity and Tissue Repair in the Lung. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:2724-2734. [PMID: 31586037 PMCID: PMC6826118 DOI: 10.4049/jimmunol.1900640] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/09/2019] [Indexed: 01/03/2023]
Abstract
Alternatively activated macrophages are essential effector cells during type 2 immunity and tissue repair following helminth infections. We previously showed that Ym1, an alternative activation marker, can drive innate IL-1R-dependent neutrophil recruitment during infection with the lung-migrating nematode, Nippostrongylus brasiliensis, suggesting a potential role for the inflammasome in the IL-1-mediated innate response to infection. Although inflammasome proteins such as NLRP3 have important proinflammatory functions in macrophages, their role during type 2 responses and repair are less defined. We therefore infected Nlrp3 -/- mice with N. brasiliensis Unexpectedly, compared with wild-type (WT) mice, infected Nlrp3 -/- mice had increased neutrophilia and eosinophilia, correlating with enhanced worm killing but at the expense of increased tissue damage and delayed lung repair. Transcriptional profiling showed that infected Nlrp3 -/- mice exhibited elevated type 2 gene expression compared with WT mice. Notably, inflammasome activation was not evident early postinfection with N. brasiliensis, and in contrast to Nlrp3 -/- mice, antihelminth responses were unaffected in caspase-1/11-deficient or WT mice treated with the NLRP3-specific inhibitor MCC950. Together these data suggest that NLRP3 has a role in constraining lung neutrophilia, helminth killing, and type 2 immune responses in an inflammasome-independent manner.
Collapse
MESH Headings
- Animals
- Caspase 1/physiology
- Chemotaxis, Leukocyte
- Eosinophilia/etiology
- Eosinophilia/immunology
- Furans/pharmacology
- Heterocyclic Compounds, 4 or More Rings
- Immunity, Innate
- Indenes
- Inflammasomes/physiology
- Interleukin-4/pharmacology
- Lectins/biosynthesis
- Lectins/genetics
- Lung/pathology
- Lung/physiology
- Lung Diseases, Parasitic/complications
- Lung Diseases, Parasitic/immunology
- Lung Diseases, Parasitic/pathology
- Lung Diseases, Parasitic/physiopathology
- Macrophages, Alveolar/enzymology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/physiology
- Neutrophils/immunology
- Nippostrongylus/immunology
- Regeneration
- Strongylida Infections/complications
- Strongylida Infections/immunology
- Strongylida Infections/pathology
- Strongylida Infections/physiopathology
- Sulfonamides/pharmacology
- Sulfones
- Transcription, Genetic
- beta-N-Acetylhexosaminidases/biosynthesis
- beta-N-Acetylhexosaminidases/genetics
Collapse
Affiliation(s)
- Alistair L Chenery
- Wellcome Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Rafid Alhallaf
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - Zainab Agha
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - Jesuthas Ajendra
- Wellcome Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - James E Parkinson
- Wellcome Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Martha M Cooper
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - Brian H K Chan
- Wellcome Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Ramon M Eichenberger
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - Lindsay A Dent
- Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5000, Australia; and
| | - Avril A B Robertson
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - David Brough
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - Tara E Sutherland
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Judith E Allen
- Wellcome Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom;
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Paul R Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia;
| |
Collapse
|
41
|
NLRP3 gain-of-function in CD4 + T lymphocytes ameliorates experimental autoimmune encephalomyelitis. Clin Sci (Lond) 2019; 133:1901-1916. [PMID: 31471462 DOI: 10.1042/cs20190506] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/27/2019] [Accepted: 08/30/2019] [Indexed: 12/21/2022]
Abstract
NLRP3 inflammasome [NLR (nucleotide-binding domain, leucine-rich repeat containing protein) Pyrin-domain-containing 3 ] functions as an innate sensor of several PAMPs and DAMPs (pathogen- and damage-associated molecular patterns). It has been also reported as a transcription factor related to Th2 pattern, although its role in the adaptive immunity has been controversial, mainly because the studies were performed using gene deletion approaches. In the present study, we have investigated the NLRP3 gain-of-function in the context of encephalomyelitis autoimmune disease (EAE), considered to be a Th1- and Th17-mediated disease. We took advantage of an animal model with NLRP3 gain-of-function exclusively to T CD4+ lymphocytes (CD4CreNLRP3fl/fl). These mice presented reduced clinical score, accompanied by less infiltrating T CD4+ cells expressing both IFN-γ and IL-17 at the central nervous system (CNS) during the peak of the disease. However, besides NLRP3 gain-of-function in lymphocytes, these mice lack NLRP3 expression in non-T CD4+ cells. Therefore, in order to circumvent this deficiency, we transferred naive CD4+ T cells from WT, NLRP3-/- or CD4CreNLRP3fl/fl into Rag-1-/- mice and immunized them with MOG35-55 Likewise, the animals repopulated with CD4CreNLRP3fl/fl T CD4+ cells presented reduced clinical score and decreased IFN-γ production at the peak of the disease. Additionally, primary effector CD4+ T cells derived from these mice presented reduced glycolytic profile, a metabolic profile compatible with Th2 cells. Finally, naive CD4+ T cells from CD4CreNLRP3fl/fl mice under a Th2-related cytokine milieu cocktail exhibited in vitro an increased IL-4 and IL-13 production. Conversely, naive CD4+ T cells from CD4CreNLRP3fl/fl mice under Th1 differentiation produced less IFN-γ and T-bet. Altogether, our data evidence that the NLRP3 gain-of-function promotes a Th2-related response, a pathway that could be better explored in the treatment of multiple sclerosis.
Collapse
|
42
|
Hrabar J, Trumbić Ž, Bočina I, Bušelić I, Vrbatović A, Mladineo I. Interplay between proinflammatory cytokines, miRNA, and tissue lesions in Anisakis-infected Sprague-Dawley rats. PLoS Negl Trop Dis 2019; 13:e0007397. [PMID: 31091271 PMCID: PMC6538193 DOI: 10.1371/journal.pntd.0007397] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 04/16/2019] [Indexed: 12/29/2022] Open
Abstract
Background Anisakiasis is an emerging public health problem, caused by Anisakis spp. nematode larvae. Anisakiasis presents as variable and unspecific gastrointestinal and/or allergic clinical symptoms, which accounts for the high rate of misdiagnosed cases. Methodology/Principal findings The aim of this study was to characterize the early cellular (6–72 h p.i.) and molecular (6 h p.i.) immune response and general underlying regulatory mechanism in Anisakis infected rats. Each Sprague-Dawley rat was infected with 10 Anisakis spp. larvae by gastric intubation. Tissues with visible lesions were processed for: i) classic histopathology (HE), immunofluorescence (CD3, iNOS, S100A8/A9), and transmission electron microscopy (TEM); ii) target genes (Il1b, Il6, Il18, Ccl3, Icam1, Mmp9) and microRNA (Rat Immunopathology MIRN-104ZF plate, Quiagen) expression analysis; and iii) global DNA methylation. Histopathology revealed that Anisakis larval migration caused moderate to extensive hemorrhages in submucosal and epimysial/perimysial connective tissue. In stomach and muscle, moderate to abundant mixed inflammatory infiltrate was present, dominated by neutrophils and macrophages, while only mild infiltration was seen in intestine. Lesions were characterized by the presence of CD3+, iNOS+, and S100A8/A9+ cells. The greatest number of iNOS+ and S100A8/A9+ cells was seen in muscle. Il6, Il1b, and Ccl3 showed particularly strong expression in stomach and visceral adipose tissues, but the order of expression differed between tissues. In total, three miRNAs were differentially expressed, two in stomach (miRNA-451 and miRNA-223) and two in intestine (miRNA-451 and miRNA-672). No changes in global DNA methylation were observed in infected tissues relative to controls. Conclusions/Significance Anisakis infection induces strong immune responses in infected rats with marked induction of specific proinflammatory cytokines and miRNA expression. Deciphering the functional role of these cytokines and miRNAs will help in understanding the anisakiasis pathology and controversies surrounding Anisakis infection in humans. Anisakiasis is a zoonotic disease (infection transmitted between animals and humans) contracted by consumption of raw or undercooked seafood contaminated with Anisakis spp. nematode larvae. Anisakiasis usually presents with variable and unspecific gastrointestinal and/or allergic symptoms, which accounts for the high rate of misdiagnosed cases. Due to changes in dietary habits, such as eating raw or undercooked seafood, anisakiasis is considered an emerging public health problem. Despite the increase in number of reported cases worldwide, mechanisms of immune response to this unspecific human pathogen are poorly known. We have shown that in experimentally infected rats, Anisakis larvae cause severe hemorrhages and necrotic changes of affected tissues in the early phase of infections. Neutrophils and macrophages were abundantly present in tissue lesions, while eosinophils, hallmark of helminth infections, were scarcely present. We have also demonstrated particularly strong expression of several inflammatory genes. Moreover, we give for the first-time insight into putative regulatory mechanism mediated via a distinct class of RNA molecules. Our study may provide new opportunities for better understanding of cellular and molecular response to Anisakis spp., aiming at development of more specific therapeutics and alleviation of pathologies associated with Anisakis spp. infection.
Collapse
Affiliation(s)
- Jerko Hrabar
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
- * E-mail:
| | - Željka Trumbić
- Department of Marine Studies, University of Split, Split, Croatia
| | - Ivana Bočina
- Faculty of Science, University of Split, Split, Croatia
| | - Ivana Bušelić
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| | - Anamarija Vrbatović
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| | - Ivona Mladineo
- Laboratory of Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| |
Collapse
|
43
|
Celias DP, Corvo I, Silvane L, Tort JF, Chiapello LS, Fresno M, Arranz A, Motrán CC, Cervi L. Cathepsin L3 From Fasciola hepatica Induces NLRP3 Inflammasome Alternative Activation in Murine Dendritic Cells. Front Immunol 2019; 10:552. [PMID: 30967874 PMCID: PMC6438957 DOI: 10.3389/fimmu.2019.00552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
The production of IL-1-family cytokines such as IL-1β and IL-18 is finely regulated by inflammasome activation after the recognition of pathogens associated molecular pattern (PAMPs) and danger associated molecular patterns (DAMPs). However, little is known about the helminth-derived molecules capable of activating the inflammasome. In the case of the helminth trematode Fasciola hepatica, the secretion of different cathepsin L cysteine peptidases (FhCL) is crucial for the parasite survival. Among these enzymes, cathepsin L3 (FhCL3) is expressed mainly in the juvenile or invasive stage. The ability of FhCL3 to digest collagen has demonstrated to be critical for intestinal tissue invasion during juvenile larvae migration. However, there is no information about the interaction of FhCL3 with the immune system. It has been shown here that FhCL3 induces a non-canonical inflammasome activation in dendritic cells (DCs), leading to IL-1β and IL-18 production without a previous microbial priming. Interestingly, this activation was depending on the cysteine protease activity of FhCL3 and the NLRP3 receptor, but independent of caspase activation. We also show that FhCL3 is internalized by DCs, promoting pro-IL-1β cleavage to its mature and biologically active form IL-1β, which is released to the extracellular environment. The FhCL3-induced NLRP3 inflammasome activation conditions DCs to promote a singular adaptive immune response, characterized by increased production of IFN-γ and IL-13. These data reveal an unexpected ability of FhCL3, a helminth-derived molecule, to activate the NLRP3 inflammasome, which is independent of the classical mechanism involving caspase activation.
Collapse
Affiliation(s)
- Daiana Pamela Celias
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Ileana Corvo
- Laboratorio de Investigación y Desarrollo de Moléculas Bioactivas, CENUR Litoral Norte - Sede Paysandú, Universidad de la República, Paysandú, Uruguay
| | - Leonardo Silvane
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - José Francisco Tort
- Departmento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laura Silvina Chiapello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa (CSIC), Madrid, Spain
| | - Alicia Arranz
- Centro de Biología Molecular Severo Ochoa (CSIC), Madrid, Spain
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| |
Collapse
|
44
|
Wu J, Li H, Xie H, Wu X, Lan P. The malignant role of exosomes in the communication among colorectal cancer cell, macrophage and microbiome. Carcinogenesis 2019; 40:601-610. [PMID: 30864655 DOI: 10.1093/carcin/bgy138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/15/2018] [Accepted: 02/27/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jinjie Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haiyan Li
- Department of Breast and Thyroid Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hongyu Xie
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xianrui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Abstract
Sepsis was known to ancient Greeks since the time of great physician Hippocrates (460-377 BC) without exact information regarding its pathogenesis. With time and medical advances, it is now considered as a condition associated with organ dysfunction occurring in the presence of systemic infection as a result of dysregulation of the immune response. Still with this advancement, we are struggling for the development of target-based therapeutic approach for the management of sepsis. The advancement in understanding the immune system and its working has led to novel discoveries in the last 50 years, including different pattern recognition receptors. Inflammasomes are also part of these novel discoveries in the field of immunology which are <20 years old in terms of their first identification. They serve as important cytosolic pattern recognition receptors required for recognizing cytosolic pathogens, and their pathogen-associated molecular patterns play an important role in the pathogenesis of sepsis. The activation of both canonical and non-canonical inflammasome signaling pathways is involved in mounting a proinflammatory immune response via regulating the generation of IL-1β, IL-18, IL-33 cytokines and pyroptosis. In addition to pathogens and their pathogen-associated molecular patterns, death/damage-associated molecular patterns and other proinflammatory molecules involved in the pathogenesis of sepsis affect inflammasomes and vice versa. Thus, the present review is mainly focused on the inflammasomes, their role in the regulation of immune response associated with sepsis, and their targeting as a novel therapeutic approach.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Australia,
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia,
| |
Collapse
|
46
|
Zakeri A, Hansen EP, Andersen SD, Williams AR, Nejsum P. Immunomodulation by Helminths: Intracellular Pathways and Extracellular Vesicles. Front Immunol 2018; 9:2349. [PMID: 30369927 PMCID: PMC6194161 DOI: 10.3389/fimmu.2018.02349] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
Helminth parasites are masters at manipulating host immune responses, using an array of sophisticated mechanisms. One of the major mechanisms enabling helminths to establish chronic infections is the targeting of pattern recognition receptors (PRRs) including toll-like receptors, C-type lectin receptors, and the inflammasome. Given the critical role of these receptors and their intracellular pathways in regulating innate inflammatory responses, and also directing adaptive immunity toward Th1 and Th2 responses, recognition of the pathways triggered and/or modulated by helminths and their products will provide detailed insights about how helminths are able to establish an immunoregulatory environment. However, helminths also target PRRs-independent mechanisms (and most likely other yet unknown mechanisms and pathways) underpinning the battery of different molecules helminths produce. Herein, the current knowledge on intracellular pathways in antigen presenting cells activated by helminth-derived biomolecules is reviewed. Furthermore, we discuss the importance of helminth-derived vesicles as a less-appreciated components released during infection, their role in activating these host intracellular pathways, and their implication in the development of new therapeutic approaches for inflammatory diseases and the possibility of designing a new generation of vaccines.
Collapse
Affiliation(s)
- Amin Zakeri
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Eline P. Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Sidsel D. Andersen
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Andrew R. Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Inclan-Rico JM, Siracusa MC. First Responders: Innate Immunity to Helminths. Trends Parasitol 2018; 34:861-880. [PMID: 30177466 PMCID: PMC6168350 DOI: 10.1016/j.pt.2018.08.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/11/2018] [Accepted: 08/12/2018] [Indexed: 02/07/2023]
Abstract
Helminth infections represent a significant public health concern resulting in devastating morbidity and economic consequences across the globe. Helminths migrate through mucosal sites causing tissue damage and the induction of type 2 immune responses. Antihelminth protection relies on the mobilization and activation of multiple immune cells, including type 2 innate lymphocytes (ILC2s), basophils, mast cells, macrophages, and hematopoietic stem/progenitor cells. Further, epithelial cells and neurons have been recognized as important regulators of type 2 immunity. Collectively, these pathways stimulate host-protective responses necessary for worm expulsion and the healing of affected tissues. In this review we focus on the innate immune pathways that regulate immunity to helminth parasites and describe how better understanding of these pathways may lead to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Mark C Siracusa
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|