1
|
Roy M, Bagchi A, Karmakar C, Chatterjee M. Transdifferentiation of neutrophils facilitates the establishment of infection by Leishmania donovani parasites. Infect Immun 2025:e0040924. [PMID: 40340446 DOI: 10.1128/iai.00409-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/10/2025] Open
Abstract
Neutrophil transdifferentiation involves the acquisition of dendritic cell-like properties, challenging the traditional view of neutrophils being solely phagocytes. The presence of transdifferentiated neutrophils is established in Visceral Leishmaniasis, but not in its dermal sequel, Post Kala-azar Dermal Leishmaniasis. Accordingly, this study investigated the altered functionalities of neutrophils focusing on the acquisition of dendritic cell-like properties and its impact on infection establishment. In PKDL cases, immunophenotyping of neutrophil-dendritic cells (N-DC hybrids) was performed using flow cytometry, along with studying the status of N-DC hybrid inducing cytokines (TNF-α, IFN-γ) and growth factor (GM-CSF). Ex vivo infection of neutrophils with L. donovani was monitored by droplet digital PCR, employing A2; additionally, their frequency of transdifferentiation, oxidative and phagocytic status, as well as apoptosis potential were quantified by flow cytometry. Compared with healthy controls, neutrophils from PKDL cases demonstrated a significant upregulation of CD83 positivity, but the frequency of co-stimulation (HLA-DR, CD80/86) was unaltered. PKDL cases demonstrated raised levels of TNF-α and IFN-γ, but GM-CSF remained unchanged. Following ex vivo infection of neutrophils, infection was evident at 2 h and was accompanied by CD83 positivity. Furthermore, the CD66b+/CD83 vis-à-vis CD66b+/CD83- subset exhibited heightened generation of reactive oxygen species (ROS), enhanced phagocytosis, and increased apoptosis. Taken together, neutrophils from PKDL cases demonstrated transdifferentiation with the absence of antigen-presenting function. Virulent Leishmania induced transdifferentiation in neutrophils, altering their functionalities and facilitating parasite uptake, along with heightened generation of intra-neutrophilic ROS and enhanced apoptosis, which possibly facilitated their engulfment by macrophages, thereby bolstering the "Trojan horse" mechanism of parasite transfer.
Collapse
Affiliation(s)
- Madhurima Roy
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Aniruddha Bagchi
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Chaitali Karmakar
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research (IPGME&R), Kolkata, India
| |
Collapse
|
2
|
Chaves MM. Neutrophils and purinergic signaling: Partners in the crime against Leishmania parasites? Biochimie 2025; 232:43-53. [PMID: 39855456 DOI: 10.1016/j.biochi.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The parasite of the genus Leishmania is the causative agent of diseases that affect humans called leishmaniasis. These diseases affect millions of people worldwide and the currently existing drugs are either very toxic or the parasites acquire resistance. Therefore, new elimination mechanisms need to be elucidated so that new therapeutic strategies can be developed. Much has already been discussed about the role of neutrophils in Leishmania infection, and their participation is still controversial. A recent study showed that receptors present in the neutrophil membrane, the purinergic receptors, can control the infection when activated, but the triggering mechanism has not been elucidated. In this review, we will address the possible participation of purinergic receptors expressed in the neutrophil extracellular membrane that may be participating in the detection of Leishmania infection and their possible effects during parasitism.
Collapse
Affiliation(s)
- Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, Brazil.
| |
Collapse
|
3
|
Verçosa BLA, Muniz-Junqueira MI, Borges LDF, Melo MN, Vasconcelos AC. Enhanced apoptosis, inflammatory cellularity, collagen deposition, and interaction between fibroblasts and Leishmania amastigotes in undamaged ear skin of dogs with leishmaniosis. Vet Parasitol 2025; 337:110488. [PMID: 40315687 DOI: 10.1016/j.vetpar.2025.110488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Fibroblasts are located close to the area of skin inoculation of Leishmania promastigotes. They are a potential cellular target for early parasite infection, harboring amastigotes of Leishmania spp. This study aimed to determine the apoptosis in fibroblasts, and to correlate these results with inflammation, parasite load, AgNOR (Argyrophilic Nucleolar Organizer Region) index, and clinical features in Leishmania-affected dogs. Fragments from the undamaged ear skin of 16 Leishmania-infected and seven uninfected dogs were evaluated by histomorphometry and immunohistochemical analysis, which correlated fibroblast apoptosis to clinical manifestation and parasite load. Ultra-thin sections were examined under transmission electronic microscopy (TEM). When applying immunohistochemical analysis, Leishmania amastigotes were only found in clinically affected dogs. The cellularity of the inflammatory infiltrate and the AgNOR index (fibroblasts and inflammatory infiltrate) were higher in clinically affected dogs. The collagen deposition score was statistically significantly higher in Leishmania-infected dogs. The apoptotic index of inflammatory cells and fibroblasts proved to be higher in clinically affected dogs. From an ultrastructural point of view, apoptotic cells shrank, while the nuclear chromatin and cytoplasm condensed. Amastigotes were observed within inflammatory cells (neutrophils and macrophages) and in the inner portions of fibroblasts. Fibroblast apoptosis was related to both the increase in the parasite load and the intensity of the inflammatory response. Histomorphometric assessments (inflammation, parasite load, AgNOR index, and apoptosis) and clinical manifestations were also associated. Collagen deposition was positively correlated with AgNOR expression and the apoptotic index (inflammatory cell and fibroblast). Therefore, fibroblast apoptosis contributes to the infection process, pathogenesis, and chronicity of canine leishmaniosis. Moreover, fibroblasts may well provide an escape mechanism for immune defenses against Leishmania.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.
| | | | - Luciano de F Borges
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil.
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
5
|
Goto Y. Immunomodulation by Leishmania parasites: Potential for controlling other diseases. Parasitol Int 2025; 104:102987. [PMID: 39515578 DOI: 10.1016/j.parint.2024.102987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
In the mammalian hosts, Leishmania parasites survive and proliferate within phagolysosomes of macrophages. To avoid being killed by the immune cells, Leishmania parasites utilize their molecules to manipulate macrophages' functions for survival. Targets of such immunomodulatory molecules are not limited to macrophages, as Leishmania-derived molecules sometimes show influence on other immune cells such as neutrophils, dendritic cells, T cells and B cells. This review covers research on immunomodulation of host immunity by Leishmania parasites and introduces some examples of parasite-derived molecules participating in the immunomodulation. For example, Leishmania cell surface lipophosphoglycan (LPG) can modulate TLR2 signaling and PI3K/Akt axis in macrophages leading to induction of Th2 cells. Because chronic secretion of inflammatory cytokines is one of the causes of immune-mediated diseases such as atherosclerosis, Crohn's disease, and rheumatoid arthritis, LPG may be useful as a drug to suppress the inflammatory conditions. The unique characteristics of leishmanial molecules pose a promise as a source of immunomodulatory drugs for controlling diseases other than leishmaniasis.
Collapse
Affiliation(s)
- Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan.
| |
Collapse
|
6
|
Verçosa BLA, Muniz-Junqueira MI, Mineiro ALBB, Melo MN, Vasconcelos AC. Enhanced apoptosis and inflammation allied with autophagic and apoptotic Leishmania amastigotes in the seemingly undamaged ear skin of clinically affected dogs with canine visceral Leishmaniasis. Cell Immunol 2025; 408:104909. [PMID: 39701006 DOI: 10.1016/j.cellimm.2024.104909] [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: 09/16/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Programmed cell death plays a relevant role in the pathogenesis of visceral Leishmaniasis. Apoptosis selects suitable parasites, regulating parasite density, whereas autophagy eliminates pathogens. This study aimed to assess the inflammation and apoptosis in inflammatory cells and presents a unique description of the presence of autophagic and apoptotic Leishmania amastigotes in naturally Leishmania-infected dogs. Fragments from seemingly undamaged ear skin of sixteen Leishmania-infected dogs and seven uninfected dogs were evaluated through histomorphometry, ultrastructural, immunohistochemical and transmission electron microscopy (TEM) analyses. Leishmania amastigotes were present on seemingly undamaged ear skin only in clinically affected dogs. Parasite load, morphometrical parameters of inflammation and apoptotic index of inflammatory cells were higher in clinically affected animals and were related to clinical manifestations. Apoptotic index and morphometric parameters of the inflammatory infiltrate in undamaged ear skin were positively correlated with parasite load. Apoptotic and non-apoptotic Leishmania amastigotes were observed within neutrophils and macrophages. Leishmania amastigotes were positive for Bax, a marker for apoptosis, by immunohistochemistry. Morphological characteristics of apoptosis and autophagy in Leishmania amastigotes were observed only in phagocytes of clinically affected dogs. Positive correlations were found between histomorphometry and clinical manifestations. Our results showed that apoptosis and autophagy in Leishmania amastigotes may be related to both the increase in parasite load and apoptotic index in inflammatory cells, and with the intensity of the inflammatory response in clinically affected dogs. Thus, our study suggests that apoptotic and autophagy Leishmania within phagocytes may have facilitate the survival of the parasite and it appears to play an important role in the process of Leishmania infection.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil; Faculdade de Ciências da Saúde Pitágoras, Campus Codó, Codó, Maranhão, Brazil.
| | | | - Ana Lys Bezerra Barradas Mineiro
- Departamento de Clínica e Cirurgia Veterinária, Centro de Ciências Agrárias, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
7
|
Luo J, Huang H, Jiang J, Zheng W, Chen P, Bai H. Clearance of Intracellular Pathogens with Hyaluronic Acid Nanomicelles Responsive to H 2S and pH. Molecules 2024; 29:5971. [PMID: 39770059 PMCID: PMC11678907 DOI: 10.3390/molecules29245971] [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: 11/07/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Hyaluronic acid (HA) is an acidic mucopolysaccharide of animal origin composed of repeating disaccharide units of N-acetylglucosamine and glucuronic acid. Due to its excellent biocompatibility, biodegradability, and selective affinity for CD44 receptors on cell surfaces, HA is widely employed as a drug carrier. In our study, we aimed to target subcellular bacteria by grafting cystamine onto HA scaffolds through an amide reaction, producing a linker responsive to H2S and pH changes. Subsequently, hydrophobic dodecylamine was attached to HA, forming amphiphilic molecules. These amphiphilic entities can self-assemble into nanomicelles in an aqueous solution, thereby encapsulating the antibacterial agent triclosan (TCS). The resulting HA-based system (HASS-TCS) can be internalized via CD44-mediated endocytosis, releasing substantial amounts of streptomycin and TCS in H2S-rich and acidic environments. Additionally, HASS-TCS has demonstrated effectiveness in eradicating biofilms and addressing intracellular infections caused by Salmonella. This study underscores a novel pH-sensitive hyaluronic acid-based drug delivery system with significant potential for the effective treatment of intracellular infections.
Collapse
Affiliation(s)
| | | | | | | | - Peng Chen
- Engineering Laboratory of Chemical Resources Utilization in South Xinjiang, Tarim University, Alar 843300, China; (J.L.); (H.H.); (J.J.); (W.Z.)
| | - Hongjin Bai
- Engineering Laboratory of Chemical Resources Utilization in South Xinjiang, Tarim University, Alar 843300, China; (J.L.); (H.H.); (J.J.); (W.Z.)
| |
Collapse
|
8
|
Teixeira TL, Teixeira SC, Borges BC, Servato JPS, Oliveira ECMD, Velikkakam T, Silva CVD. Trypanosoma cruzi P21 protein exacerbates Leishmania (L.) amazonensis infection. Exp Parasitol 2024; 267:108856. [PMID: 39461608 DOI: 10.1016/j.exppara.2024.108856] [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: 03/26/2024] [Revised: 09/26/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
The protozoan parasite Trypanosoma cruzi, the etiological agent of Chagas disease, affects millions of people worldwide. Current treatments rely on drugs effective only in the acute phase, making the search for new therapeutic targets a priority. While a recombinant protein based on T. cruzi P21 (rP21) exhibits immunomodulatory properties and contributes to controlling parasitism and inflammation during T. cruzi infection, its efficacy against other trypanosomatids remains unexplored. This study investigated the impact of rP21 on Leishmania (L.) amazonensis infection in a murine model. Contrary to our expectations, treatment with rP21 did not ameliorate L. (L.) amazonensis infection. Instead, rP21 treatment resulted in increased parasite load in the paws of infected BALB/c mice, evidenced by larger lesion sizes and higher parasite burdens, accompanied by an intensified inflammatory infiltrate in the paw tissue. These findings suggest that despite its promising effects in the context of T. cruzi infection, rP21 may not be a suitable therapeutic candidate for L. amazonensis infection and might even exacerbate disease.
Collapse
Affiliation(s)
- Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | | | | | - Teresiama Velikkakam
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
9
|
Roy M, Sengupta R, Chakraborty BC, Chatterjee U, von Stebut E, Kaye PM, Chatterjee M. Role of neutrophils in the pathogenesis of Post Kala-azar Dermal Leishmaniasis (PKDL). PLoS Negl Trop Dis 2024; 18:e0012655. [PMID: 39602398 PMCID: PMC11602034 DOI: 10.1371/journal.pntd.0012655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Post Kala-azar Dermal Leishmaniasis (PKDL) is a dermal sequel of visceral leishmaniasis (VL), poses a significant threat to the success of ongoing kala-azar elimination program, due to its potential role in sustaining transmission cycles and complicating disease management strategies. In VL, neutrophils have been identified as the 'first line of defence', having multiple roles in disease pathogenesis, but their role in PKDL, if any, still remains elusive; presenting a critical gap in knowledge, and was the aim of this study. METHODOLOGY/PRINCIPAL FINDINGS In a cohort of PKDL patients, CD66b+ neutrophils were quantified in skin biopsies, followed by immunostaining of FFPE sections to identify activated neutrophils (CD66b+/CD64+) and degranulated (CD66b+/MPO+), along with expression of neutrophil elastase (NE), matrix metalloprotease 9 (MMP9) and collagen I. Plasma levels of neutrophil chemo-attractants CXCL8/1/2/5, CCL2 and 20 and cytokines, (IL-6, IFN-γ, IL-4, IL-10, TNF-α, IL-17 and IL-22, 23) were evaluated by a multiplex assay, while lesional expression of IL-8, IL-10 and IL-17 was evaluated by immunohistochemistry. As compared to healthy individuals (control skin samples), PKDL cases at the lesional sites had an increased number of activated CD66b+ neutrophils (positive for CD64+, MPO+ and NE+). The plasma levels of neutrophil chemo-attractants, pro-inflammatory and regulatory cytokines were raised as was circulating and lesional IL-8, along with an enhanced lesional expression of IL-10 and IL-17A. An increase in circulatory and lesional MMP9 was accompanied by decreased collagen I, suggesting disintegration of matrix integrity. CONCLUSIONS/SIGNIFICANCE Taken together, in PKDL, activated neutrophils possibly contribute towards modulating the lesional landscape. Understanding this involvement of neutrophils in patients with PKDL, particularly in the absence of an animal model, could offer better understanding of the disease pathogenesis and provide insights into novel therapeutic strategies for the ongoing elimination program.
Collapse
Affiliation(s)
- Madhurima Roy
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Ritika Sengupta
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Bidhan Chandra Chakraborty
- Multidisciplinary Research Unit (MRU) Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Uttara Chatterjee
- Pathology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Esther von Stebut
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Mitali Chatterjee
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| |
Collapse
|
10
|
DeSouza-Vieira T, Pretti MAM, Lima Gomes PS, Paula-Neto HA, Goundry A, Nascimento MT, Ganesan S, Gonçalves da Silva T, Kamenyeva O, Kabat J, Manzella-Lapeira J, B. Canto F, Fraga-Junior VDS, Eustáquio Lopes M, Gomes Vaz L, Pessenda G, Paun A, Freitas-Mesquita AL, Meyer-Fernandes JR, Boroni M, Bellio M, Batista Menezes G, Brzostowski J, Mottram J, Sacks D, Lima APCA, Saraiva EM. Functional plasticity shapes neutrophil response to Leishmania major infection in susceptible and resistant strains of mice. PLoS Pathog 2024; 20:e1012592. [PMID: 39378227 PMCID: PMC11488723 DOI: 10.1371/journal.ppat.1012592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
Neutrophils rapidly infiltrate sites of infection and possess several microbicidal strategies, such as neutrophil extracellular traps release and phagocytosis. Enhanced neutrophil infiltration is associated with higher susceptibility to Leishmania infection, but neutrophil effector response contribution to this phenotype is uncertain. Here, we show that neutrophils from susceptible BALB/c mice (B/c) produce more NETs in response to Leishmania major than those from resistant C57BL/6 mice (B6), which are more phagocytic. The absence of neutrophil elastase contributes to phagocytosis regulation. Microarray analysis shows enrichment of genes involved in NET formation (mpo, pi3kcg, il1b) in B/c, while B6 shows upregulation of genes involved in phagocytosis and cell death (Arhgap12, casp9, mlkl, FasL). scRNA-seq in L. major-infected B6 showed heterogeneity in the pool of intralesional neutrophils, and we identified the N1 subset as the putative subpopulation involved with phagocytosis. In vivo, imaging validates NET formation in infected B/c ears where NETing neutrophils were mainly uninfected cells. NET digestion in vivo augmented parasite lymphatic drainage. Hence, a balance between NET formation and phagocytosis in neutrophils may contribute to the divergent phenotype observed in these mice.
Collapse
Affiliation(s)
- Thiago DeSouza-Vieira
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marco Antônio M. Pretti
- Laboratório de Bioinformática e Biologia Computacional, Divisão de Pesquisa Experimental Translacional, Instituto Nacional do Câncer (INCA), Rio de Janeiro, Brasil
| | - Phillipe Souza Lima Gomes
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Heitor A. Paula-Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Amy Goundry
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Michelle T. Nascimento
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Triciana Gonçalves da Silva
- National Center for Structural Biology and Bioimaging, CENABIO, Universidade Federal do Rio de Janeiro, Brazil
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Javier Manzella-Lapeira
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Fábio B. Canto
- Laboratório de Tolerância Imunológica e Homeostase Linfocitária, Departamento de Imunobiologia, Universidade Federal Fluminense, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Vanderlei da Silva Fraga-Junior
- Laboratório de Imunologia Molecular e Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Mateus Eustáquio Lopes
- Centro de Biologia Gastrointestinal, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Leonardo Gomes Vaz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte, Minas Gerais, Brasil
| | - Gabriela Pessenda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anita L. Freitas-Mesquita
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Mariana Boroni
- Laboratório de Bioinformática e Biologia Computacional, Divisão de Pesquisa Experimental Translacional, Instituto Nacional do Câncer (INCA), Rio de Janeiro, Brasil
| | - Maria Bellio
- Laboratório de Imunobiologia, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Gustavo Batista Menezes
- Centro de Biologia Gastrointestinal, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Joseph Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jeremy Mottram
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ana Paula C. A. Lima
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Elvira M. Saraiva
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
11
|
Goris M, Passelli K, Peyvandi S, Díaz-Varela M, Billion O, Prat-Luri B, Demarco B, Desponds C, Termote M, Iniguez E, Dey S, Malissen B, Kamhawi S, Hurrell BP, Broz P, Tacchini-Cottier F. NLRP1-dependent activation of Gasdermin D in neutrophils controls cutaneous leishmaniasis. PLoS Pathog 2024; 20:e1012527. [PMID: 39250503 PMCID: PMC11412672 DOI: 10.1371/journal.ppat.1012527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Intracellular pathogens that replicate in host myeloid cells have devised ways to inhibit the cell's killing machinery. Pyroptosis is one of the host strategies used to reduce the pathogen replicating niche and thereby control its expansion. The intracellular Leishmania parasites can survive and use neutrophils as a silent entry niche, favoring subsequent parasite dissemination into the host. Here, we show that Leishmania mexicana induces NLRP1- and caspase-1-dependent Gasdermin D (GSDMD)-mediated pyroptosis in neutrophils, a process critical to control the parasite-induced pathology. In the absence of GSDMD, we observe an increased number of infected dermal neutrophils two days post-infection. Using adoptive neutrophil transfer in neutropenic mice, we show that pyroptosis contributes to the regulation of the neutrophil niche early after infection. The critical role of neutrophil pyroptosis and its positive influence on the regulation of the disease outcome was further demonstrated following infection of mice with neutrophil-specific deletion of GSDMD. Thus, our study establishes neutrophil pyroptosis as a critical regulator of leishmaniasis pathology.
Collapse
Affiliation(s)
- Michiel Goris
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Katiuska Passelli
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Sanam Peyvandi
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Miriam Díaz-Varela
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Oaklyne Billion
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Borja Prat-Luri
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Benjamin Demarco
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Manon Termote
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Somaditya Dey
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Post Graduate Department of Zoology, Barasat Government College, Barasat, West Bengal, India
| | - Bernard Malissen
- INSERM, CNRS, Centre D’Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Benjamin P. Hurrell
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
12
|
da Silva Lira Filho A, Lafleur A, Alvarez F, Piccirillo CA, Olivier M. Implication of the Annexin 1/FPR axis in leishmanial exosome-mediated Leishmania major skin hyperpathogenesis. Front Immunol 2024; 15:1436151. [PMID: 39076982 PMCID: PMC11284082 DOI: 10.3389/fimmu.2024.1436151] [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: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Exosomes produced by the protozoan parasite Leishmania (LeishEXO) are well-established drivers of virulence, though mechanisms underlying their exacerbation of experimental leishmaniasis remain elusive. Expression of Annexin A1 (ANXA1), a protein implicated in exosome-mediated pathologies and viral internalization, has been shown to correlate with cutaneous leishmaniasis severity. Given ANXA1's regulation of myeloid cells - the canonical hosts for Leishmania - we studied the potential role of ANXA1 and its receptors FPR1/2 in exerting LeishEXO's effects. Methods Murine and in vitro ANXA1-/- models were used to study the generation of protective TH1 responses during experimental L. major infection with and without LeishEXO. Recruitment of inflammatory cells was assessed using a peritoneal cell recruitment assay and immunophenotyping, and production of inflammatory mediators was measured using a cytokine and chemokine array. Treatment of experimental models with FPR2 antagonist WRW4 and FPR1/2 agonist WKYMVm was used to delineate the role of the FPR/ANXA1 axis in LeishEXO-mediated hyperpathogenesis. Results We established that ANXA1 deficiency prohibits LeishEXO-mediated pathogenesis and myeloid cell infection, with minimal alterations to adaptive and innate immune phenotypes. FPR2 blockade with WRW4 similarly inhibited leishmanial hyperpathogenesis, while direct activation of FPRs with WKYMVm enhanced infection and recapitulated the LeishEXO-mediated phenotype. This research describes LeishEXO's utilization of the ANXA1/FPR axis to facilitate parasitic internalization and pathogenesis, which may be leveraged in the development of therapeutics for leishmaniasis.
Collapse
Affiliation(s)
- Alonso da Silva Lira Filho
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Andrea Lafleur
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
13
|
Scaramele NF, Troiano JA, Felix JDS, Costa SF, Almeida MC, Florencio de Athayde FR, Soares MF, Lopes MFDS, Furlan ADO, de Lima VMF, Lopes FL. Leishmania infantum infection modulates messenger RNA, microRNA and long non-coding RNA expression in human neutrophils in vitro. PLoS Negl Trop Dis 2024; 18:e0012318. [PMID: 39028711 PMCID: PMC11259272 DOI: 10.1371/journal.pntd.0012318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/25/2024] [Indexed: 07/21/2024] Open
Abstract
In the Americas, L. infantum (syn. chagasi) is the main cause of human visceral leishmaniasis. The role of neutrophils as part of the innate response to Leishmania spp. infection is dubious and varies according to the species causing the infection. Global expression of coding RNAs, microRNAs and long non-coding RNAs changes as part of the immune response against pathogens. Changes in mRNA and non-coding RNA expression resulting from infection by Leishmania spp. are widely studied in macrophages, but scarce in neutrophils, the first cell to encounter the trypanosomatid, especially following infection by L. infantum. Herein, we aimed to understand the expression patterns of coding and non-coding transcripts during acute in vitro infection of human neutrophils by L. infantum. We isolated neutrophils from whole blood of healthy male donors (n = 5) and split into groups: 1) infected with L. infantum (MOI = 5:1), and 2) uninfected controls. After 3 hours of exposure of infected group to promastigotes of L. infantum, followed by 17 hours of incubation, total RNA was extracted and total RNA-Seq and miRNA microarray were performed. A total of 212 genes were differentially expressed in neutrophils following RNA-Seq analysis (log2(FC)±0.58, FDR≤0.05). In vitro infection with L. infantum upregulated the expression of 197 and reduced the expression of 92 miRNAs in human neutrophils (FC±2, FDR≤0.01). Lastly, 5 downregulated genes were classified as lncRNA, and of the 10 upregulated genes, there was only 1 lncRNA. Further bioinformatic analysis indicated that changes in the transcriptome and microtranscriptome of neutrophils, following in vitro infection with L. infantum, may impair phagocytosis, apoptosis and decrease nitric oxide production. Our work sheds light on several mechanisms used by L. infantum to control neutrophil-mediated immune response and identifies several targets for future functional studies, aiming at the development of preventive or curative treatments for this prevalent zoonosis.
Collapse
Affiliation(s)
- Natália Francisco Scaramele
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Jéssica Antonini Troiano
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Juliana de Souza Felix
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Sidnei Ferro Costa
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Mariana Cordeiro Almeida
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flávia Regina Florencio de Athayde
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Matheus Fujimura Soares
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Maria Fernanda da Silva Lopes
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Amanda de Oliveira Furlan
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flavia Lombardi Lopes
- Department of Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| |
Collapse
|
14
|
Morales-Primo AU, Becker I, Pedraza-Zamora CP, Zamora-Chimal J. Th17 Cell and Inflammatory Infiltrate Interactions in Cutaneous Leishmaniasis: Unraveling Immunopathogenic Mechanisms. Immune Netw 2024; 24:e14. [PMID: 38725676 PMCID: PMC11076297 DOI: 10.4110/in.2024.24.e14] [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: 01/14/2024] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 05/12/2024] Open
Abstract
The inflammatory response during cutaneous leishmaniasis (CL) involves immune and non-immune cell cooperation to contain and eliminate Leishmania parasites. The orchestration of these responses is coordinated primarily by CD4+ T cells; however, the disease outcome depends on the Th cell predominant phenotype. Although Th1 and Th2 phenotypes are the most addressed as steers for the resolution or perpetuation of the disease, Th17 cell activities, especially IL-17 release, are recognized to be vital during CL development. Th17 cells perform vital functions during both acute and chronic phases of CL. Overall, Th17 cells induce the migration of phagocytes (neutrophils, macrophages) to the infection site and CD8+ T cells and NK cell activation. They also provoke granzyme and perforin secretion from CD8+ T cells, macrophage differentiation towards an M2 phenotype, and expansion of B and Treg cells. Likewise, immune cells from the inflammatory infiltrate have modulatory activities over Th17 cells involving their differentiation from naive CD4+ T cells and further expansion by generating a microenvironment rich in optimal cytokines such as IL-1β, TGF-β, IL-6, and IL-21. Th17 cell activities and synergies are crucial for the resistance of the infection during the early and acute stages; however, if unchecked, Th17 cells might lead to a chronic stage. This review discusses the synergies between Th17 cells and the inflammatory infiltrate and how these interactions might destine the course of CL.
Collapse
Affiliation(s)
- Abraham U. Morales-Primo
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| | - Ingeborg Becker
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| | - Claudia Patricia Pedraza-Zamora
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, México
| | - Jaime Zamora-Chimal
- Laboratorio de Inmunoparasitología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City 06720, México
| |
Collapse
|
15
|
Teixeira SC, Teixeira TL, Tavares PCB, Alves RN, da Silva AA, Borges BC, Martins FA, Dos Santos MA, de Castilhos P, E Silva Brígido RT, Notário AFO, Silveira ACA, da Silva CV. Subversion strategies of lysosomal killing by intracellular pathogens. Microbiol Res 2023; 277:127503. [PMID: 37748260 DOI: 10.1016/j.micres.2023.127503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 09/27/2023]
Abstract
Many pathogenic organisms need to reach either an intracellular compartment or the cytoplasm of a target cell for their survival, replication or immune system evasion. Intracellular pathogens frequently penetrate into the cell through the endocytic and phagocytic pathways (clathrin-mediated endocytosis, phagocytosis and macropinocytosis) that culminates in fusion with lysosomes. However, several mechanisms are triggered by pathogenic microorganisms - protozoan, bacteria, virus and fungus - to avoid destruction by lysosome fusion, such as rupture of the phagosome and thereby release into the cytoplasm, avoidance of autophagy, delaying in both phagolysosome biogenesis and phagosomal maturation and survival/replication inside the phagolysosome. Here we reviewed the main data dealing with phagosome maturation and evasion from lysosomal killing by different bacteria, protozoa, fungi and virus.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Aline Alves da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Flávia Alves Martins
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marlus Alves Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Patrícia de Castilhos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | | | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
16
|
Bai S, Song J, Pu H, Yu Y, Song W, Chen Z, Wang M, Campbell-Valois FX, Wong WL, Cai Q, Wan M, Zhang C, Bai Y, Feng X. Chemical Biology Approach to Reveal the Importance of Precise Subcellular Targeting for Intracellular Staphylococcus aureus Eradication. J Am Chem Soc 2023; 145:23372-23384. [PMID: 37838963 DOI: 10.1021/jacs.3c09587] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Intracellular bacterial pathogens, such as Staphylococcus aureus, that may hide in intracellular vacuoles represent the most significant manifestation of bacterial persistence. They are critically associated with chronic infections and antibiotic resistance, as conventional antibiotics are ineffective against such intracellular persisters due to permeability issues and mechanistic reasons. Direct subcellular targeting of S. aureus vacuoles suggests an explicit opportunity for the eradication of these persisters, but a comprehensive understanding of the chemical biology nature and significance of precise S. aureus vacuole targeting remains limited. Here, we report an oligoguanidine-based peptidomimetic that effectively targets and eradicates intracellular S. aureus persisters in the phagolysosome lumen, and this oligomer was utilized to reveal the mechanistic insights linking precise targeting to intracellular antimicrobial efficacy. The oligomer has high cellular uptake via a receptor-mediated endocytosis pathway and colocalizes with S. aureus persisters in phagolysosomes as a result of endosome-lysosome interconversion and lysosome-phagosome fusion. Moreover, the observation of a bacterium's altered susceptibility to the oligomer following a modification in its intracellular localization offers direct evidence of the critical importance of precise intracellular targeting. In addition, eradication of intracellular S. aureus persisters was achieved by the oligomer's membrane/DNA dual-targeting mechanism of action; therefore, its effectiveness is not hampered by the hibernation state of the persisters. Such precise subcellular targeting of S. aureus vacuoles also increases the agent's biocompatibility by minimizing its interaction with other organelles, endowing excellent in vivo bacterial targeting and therapeutic efficacy in animal models.
Collapse
Affiliation(s)
- Silei Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Junfeng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel NanoOptoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China
- Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China
| | - Yue Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Wenwen Song
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Zhiyong Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | | | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong SAR, China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yugang Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
17
|
He I, Poirier B, Jensen E, Kaur S, Hedges J, Jesudason S, Jamieson L, Sethi S. Demystifying the connection between periodontal disease and chronic kidney disease - An umbrella review. J Periodontal Res 2023; 58:874-892. [PMID: 37477165 DOI: 10.1111/jre.13161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/07/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023]
Abstract
Chronic kidney disease (CKD) and poor oral health are inter-related and their significant impact on each other is well established in the literature. Many systematic reviews and meta-analyses have demonstrated a strong relationship between CKD and periodontitis, where periodontal treatment has shown potential in improving CKD outcomes. However, the quality of the studies and heterogeneity of the results show variation. The aim of this umbrella review was to review the quality of the current systematic reviews on the relationship between CKD and oral health with an emphasis on periodontal disease and to generate clinically relevant guidelines to maintain periodontal health in patients with CKD. This umbrella review was conducted and reported in alignment with the Joanna Briggs Institute and the PRISMA 2020 guidelines. The review protocol was established prior to commencing the review and registered on JBI and PROSPERO (CRD42022335209). Search strings were established for PubMed, Embase, Web of Science, Cochrane Database of Systematic Reviews, and Dentistry & Oral Science Source up to April 2022. All systematic reviews and meta-analyses that considered the relationship between CKD and periodontitis or periodontal treatment were included. Of 371 studies identified through the systematic search, 18 systematic reviews met the inclusion criteria. Ten studies assessed the relationship between oral health status and CKD with a focus on periodontitis and CKD, five reviewed the impact of periodontal treatment on CKD outcomes, two included both relationship and effectiveness of periodontal treatment and one qualitatively reviewed oral health-related quality of life in patients with kidney failure. Findings indicate there is a bidirectional relationship between CKD and periodontal disease. In view of the heterogeneity of the existing literature on CKD and periodontal disease, specific recommendations for the management of periodontitis among patients with CKD are proposed for medical professionals, dental professionals, and aged care workers based on the evidence collated in this review.
Collapse
Affiliation(s)
- Isaac He
- Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| | - Brianna Poirier
- Australian Research Centre for Population Oral Health, Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| | - Emilija Jensen
- Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sushil Kaur
- Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| | - Joanne Hedges
- Australian Research Centre for Population Oral Health, Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| | - Shilpanjali Jesudason
- Central and Northern Adelaide Renal and Transplantation Services (CNARTS), Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Lisa Jamieson
- Australian Research Centre for Population Oral Health, Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sneha Sethi
- Australian Research Centre for Population Oral Health, Adelaide Dental School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Banerjee S, Datta R. Localized Leishmania major infection disrupts systemic iron homeostasis that can be controlled by oral iron supplementation. J Biol Chem 2023; 299:105064. [PMID: 37468101 PMCID: PMC10448173 DOI: 10.1016/j.jbc.2023.105064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023] Open
Abstract
Leishmania parasites are heavily dependent on efficient iron acquisition from a tightly regulated host iron pool for survival and virulence. Prior studies uncovered multiple strategies adopted by the parasite to hijack the iron-regulatory network of macrophages. Despite these extensive studies with infected macrophages, there is limited knowledge of the effect of Leishmania infection on systemic iron homeostasis. This issue is particularly relevant for Leishmania major, which causes localized skin infection with minimal lymphatic spread. We show for the first time that L. major infection in the mouse footpad induced influx of iron at the site of infection through blood with simultaneous upregulation of transferrin receptor 1 and downregulation of phagolysosomal iron exporter Nramp1 expression in the footpad tissue. Interestingly, localized L. major infection had far-reaching effects beyond the infection site triggering anemia-like symptoms. This was evident from depleted physiological iron stores from the liver and bone marrow as well as reduced hemoglobin levels and deformed erythrocytes. The infected mice also developed splenomegaly with signs of splenic stress erythropoiesis as indicated by upregulation of several erythroid-related genes. These observations prompted us to provide oral iron supplementations to the L. major-infected mice, which resulted in a drastic reduction of the parasite load and restoration of iron homeostasis.
Collapse
Affiliation(s)
- Sourav Banerjee
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur, West Bengal, India
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur, West Bengal, India.
| |
Collapse
|
19
|
Tsioumpekou M, Krijgsman D, Leusen JHW, Olofsen PA. The Role of Cytokines in Neutrophil Development, Tissue Homing, Function and Plasticity in Health and Disease. Cells 2023; 12:1981. [PMID: 37566060 PMCID: PMC10417597 DOI: 10.3390/cells12151981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023] Open
Abstract
Neutrophils are crucial innate immune cells and comprise 50-70% of the white blood cell population under homeostatic conditions. Upon infection and in cancer, blood neutrophil numbers significantly increase because of the secretion of various chemo- and cytokines by, e.g., leukocytes, pericytes, fibroblasts and endothelial cells present in the inflamed tissue or in the tumor microenvironment (TME). The function of neutrophils in cancer has recently gained considerable attention, as they can exert both pro- and anti-tumorigenic functions, dependent on the cytokine milieu present in the TME. Here, we review the effect of cytokines on neutrophil development, tissue homing, function and plasticity in cancer and autoimmune diseases as well as under physiological conditions in the bone marrow, bloodstream and various organs like the spleen, kidney, liver, lung and lymph nodes. In addition, we address several promising therapeutic options, such as cytokine therapy, immunocytokines and immunotherapy, which aim to exploit the anti-tumorigenic potential of neutrophils in cancer treatment or block excessive neutrophil-mediated inflammation in autoimmune diseases.
Collapse
Affiliation(s)
- Maria Tsioumpekou
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
| | - Daniëlle Krijgsman
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
- Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
| | - Patricia A. Olofsen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (M.T.); (D.K.); (J.H.W.L.)
| |
Collapse
|
20
|
Ellett F, Kacamak NI, Alvarez CR, Oliveira EH, Hasturk H, Paster BJ, Kantarci A, Irimia D. Fusobacterium nucleatum dissemination by neutrophils. J Oral Microbiol 2023; 15:2217067. [PMID: 37283724 PMCID: PMC10240972 DOI: 10.1080/20002297.2023.2217067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/08/2023] Open
Abstract
Recent studies uncovered that Fusobacterium nucleatum (Fn), a common, opportunistic bacterium in the oral cavity, is associated with a growing number of systemic diseases, ranging from colon cancer to Alzheimer's disease. However, the pathological mechanisms responsible for this association are still poorly understood. Here, we leverage recent technological advances to study the interactions between Fn and neutrophils. We show that Fn survives within human neutrophils after phagocytosis. Using in vitro microfluidic devices, we determine that human neutrophils can protect and transport Fn over large distances. Moreover, we validate these observations in vivo by showing that neutrophils disseminate Fn using a zebrafish model. Our data support the emerging hypothesis that bacterial dissemination by neutrophils is a mechanistic link between oral and systemic diseases. Furthermore, our results may ultimately lead to therapeutic approaches that target specific host-bacteria interactions, including the dissemination process.
Collapse
Affiliation(s)
- Felix Ellett
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, USA
- Shriners Hospital for Children, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nazli I. Kacamak
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Carla R. Alvarez
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Eduardo H.S. Oliveira
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Hatice Hasturk
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Bruce J. Paster
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Alpdogan Kantarci
- The Forsyth Institute, Cambridge, MA, USA
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Daniel Irimia
- Department of Surgery, Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, USA
- Shriners Hospital for Children, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
de Carvalho TP, da Silva LA, Castanheira TLL, de Souza TD, da Paixão TA, Lazaro-Anton L, Tsolis RM, Santos RL. Cell and Tissue Tropism of Brucella spp. Infect Immun 2023; 91:e0006223. [PMID: 37129522 PMCID: PMC10187126 DOI: 10.1128/iai.00062-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Brucella spp. are facultatively intracellular bacteria that can infect, survive, and multiply in various host cell types in vivo and/or in vitro. The genus Brucella has markedly expanded in recent years with the identification of novel species and hosts, which has revealed additional information about the cell and tissue tropism of these pathogens. Classically, Brucella spp. are considered to have tropism for organs that contain large populations of phagocytes such as lymph nodes, spleen, and liver, as well as for organs of the genital system, including the uterus, epididymis, testis, and placenta. However, experimental infections of several different cultured cell types indicate that Brucella may actually have a broader cell tropism than previously thought. Indeed, recent studies indicate that certain Brucella species in particular hosts may display a pantropic distribution in vivo. This review discusses the available knowledge on cell and tissue tropism of Brucella spp. in natural infections of various host species, as well as in experimental animal models and cultured cells.
Collapse
Affiliation(s)
- Thaynara Parente de Carvalho
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Laice Alves da Silva
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Larissa Lourenço Castanheira
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Federal de Educação Ciência e Tecnologia do Norte de Minas Gerais, Salinas, Brazil
| | - Tayse Domingues de Souza
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tatiane Alves da Paixão
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leticia Lazaro-Anton
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Renee M. Tsolis
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Renato Lima Santos
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| |
Collapse
|
22
|
Soto-Olguín N, Zamora-Chimal J, Delgado-Domínguez J, Becker I. Leishmania mexicana Lipophosphoglycan Activates Dermal γδ T Cells with Participation of TLR2. Acta Parasitol 2023; 68:122-129. [PMID: 36434381 DOI: 10.1007/s11686-022-00639-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 11/03/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE Leishmania transmission by sand flies is detected by dermal cells that recognize ligands, such as lipophosphoglycan (LPG) on the promastigote glycocalyx. Resident dermal cells include γδ T cells, that recognize antigens by TCR or innate receptors, such as TLRs. We analyzed the response of dermal γδ T cells to Leishmania mexicana infections or inoculation of LPG, and whether parasite LPG activates γδ T cells through TLR2. METHODS We stimulated γδ T cells with LPG and analyzed colocalization of LPG and TLR2 by confocal microscopy. Activation of TLR2 was evaluated by IκBα phosphorylation. BALB/c mice were inoculated with L. mexicana or LPG in the dermis of earlobes, and LPG+ TLR2+ γδ T cells were analyzed by flow cytometry. TNF+ γδ T cells were examined in earlobe dermis by confocal microscopy. RESULTS Stimulation with purified LPG showed activation of TLR2 with IκBα phosphorylation in γδ T cells. Inoculation of L. mexicana parasites or LPG into earlobe dermis showed co-expression of LPG+ and TLR2+ in γδ T cells, demonstrating their interaction during infections. A subset of γδ T cells (LPG+ and TLR2-) provided evidence that additional receptors recognize LPG. Inoculation of LPG enhanced overall γδ T cell numbers, including those expressing TNF, whereas infection with the parasite mostly enhanced γδ T cells expressing TNF. CONCLUSION L. mexicana LPG is a ligand recognized by TLR2 on γδ-T cells leading to their activation, although contribution of other receptors cannot be ruled out and need to be analyzed to elucidate their contribution during Leishmania infections.
Collapse
Affiliation(s)
- Nadia Soto-Olguín
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, 06726, Mexico City, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, 06726, Mexico City, Mexico
| | - José Delgado-Domínguez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, 06726, Mexico City, Mexico
| | - Ingeborg Becker
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, 06726, Mexico City, Mexico.
| |
Collapse
|
23
|
Osorio EY, Uscanga-Palomeque A, Patterson GT, Cordova E, Travi BL, Soong L, Melby PC. Malnutrition-related parasite dissemination from the skin in visceral leishmaniasis is driven by PGE2-mediated amplification of CCR7-related trafficking of infected inflammatory monocytes. PLoS Negl Trop Dis 2023; 17:e0011040. [PMID: 36630476 PMCID: PMC9873180 DOI: 10.1371/journal.pntd.0011040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/24/2023] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
People are infected with Leishmania donovani when the parasite is deposited in the dermis during the blood meal of the sand fly vector. Most infected people develop a subclinical latent infection, but some develop progressive visceral leishmaniasis. Malnutrition is a risk factor for the development of active VL. We previously demonstrated increased parasite dissemination from the skin to visceral organs in a murine model of malnutrition. Here we investigated the mechanism of early parasite dissemination. After delivery of L. donovani to the skin, we found enhanced capture of parasites by inflammatory monocytes and neutrophils in the skin of malnourished mice. However, parasite dissemination in malnourished mice was driven primarily by infected inflammatory monocytes, which showed increased CCR7 expression, greater intrinsic migratory capacity, and increased trafficking from skin to spleen. PGE2 production, which was increased at the site of skin infection, increased monocyte CCR7 expression and promoted CCR7-related monocyte-mediated early parasite dissemination in malnourished mice. Parasite dissemination in monocytes was reduced by inhibition of PGE2, knockdown or silencing of CCR7 in monocytes, and depletion of inflammatory monocytes through administration of diphtheria toxin to CSFR1-DTR transgenic mice that have monocyte-specific DT receptor expression. CCR7-driven trafficking of infected inflammatory monocytes through the lymph node was accompanied by increased expression of its ligands CCL19 and CCL21. These results show that the CCR7/PGE2 axis is responsible for the increased trafficking of L. donovani-infected inflammatory monocytes from the skin to the spleen in the malnourished host. Undernutrition and production of PGE2 are potential targets to reduce the risk of people developing VL. Nutritional interventions that target improved immune function and reduced PGE2 synthesis should be studied in people at risk of developing VL.
Collapse
Affiliation(s)
- E. Yaneth Osorio
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (PCM); (EYO)
| | - Ashanti Uscanga-Palomeque
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Grace T. Patterson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Erika Cordova
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bruno L. Travi
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Peter C. Melby
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (PCM); (EYO)
| |
Collapse
|
24
|
Feng W, Chittò M, Moriarty TF, Li G, Wang X. Targeted Drug Delivery Systems for Eliminating Intracellular Bacteria. Macromol Biosci 2023; 23:e2200311. [PMID: 36189899 DOI: 10.1002/mabi.202200311] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Indexed: 01/19/2023]
Abstract
The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.
Collapse
Affiliation(s)
- Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,AO Research Institute Davos, Davos, 7270, Switzerland
| | - Marco Chittò
- AO Research Institute Davos, Davos, 7270, Switzerland
| | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
25
|
Janžič L, Repas J, Pavlin M, Zemljić-Jokhadar Š, Ihan A, Kopitar AN. Macrophage polarization during Streptococcus agalactiae infection is isolate specific. Front Microbiol 2023; 14:1186087. [PMID: 37213504 PMCID: PMC10192866 DOI: 10.3389/fmicb.2023.1186087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Streptococcus agalactiae (Group B Streptococcus, GBS), a Gram-positive commensal in healthy adults, remains a major cause of neonatal infections, usually manifesting as sepsis, meningitis, or pneumonia. Intrapartum antibiotic prophylaxis has greatly reduced the incidence of early-onset disease. However, given the lack of effective measures to prevent the risk of late-onset disease and invasive infections in immunocompromised individuals, more studies investigating the GBS-associated pathogenesis and the interplay between bacteria and host immune system are needed. Methods Here, we examined the impact of 12 previously genotyped GBS isolates belonging to different serotypes and sequence types on the immune response of THP-1 macrophages. Results Flow cytometry analysis showed isolate-specific differences in phagocytic uptake, ranging from 10% for isolates of serotype Ib, which possess the virulence factor protein β, to over 70% for isolates of serotype III. Different isolates also induced differential expression of co-stimulatory molecules and scavenger receptors with colonizing isolates inducing higher expression levels of CD80 and CD86 compared to invasive isolates. In addition, real-time measurements of metabolism revealed that macrophages enhanced both glycolysis and mitochondrial respiration after GBS infection, with isolates of serotype III being the most potent activators of glycolysis and glycolytic ATP production. Macrophages also showed differential resistance to GBS-mediated cell cytotoxicity as measured by LDH release and real-time microscopy. The differences were evident both between serotypes and between isolates obtained from different specimens (colonizing or invasive isolates) demonstrating the higher cytotoxicity of vaginal compared with blood isolates. Conclusions Thus, the data suggest that GBS isolates differ in their potential to become invasive or remain colonizing. In addition, colonizing isolates appear to be more cytotoxic, whereas invasive isolates appear to exploit macrophages to their advantage, avoiding the immune recognition and antibiotics.
Collapse
Affiliation(s)
- Larisa Janžič
- Department of Cell Immunology, Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| | - Špela Zemljić-Jokhadar
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Department of Cell Immunology, Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andreja Nataša Kopitar
- Department of Cell Immunology, Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Andreja Nataša Kopitar,
| |
Collapse
|
26
|
Liang Y, Wang H, Gonzales C, Thiriot J, Sunyakumthorn P, Melby PC, Sun J, Soong L. CCR7/dendritic cell axis mediates early bacterial dissemination in Orientia tsutsugamushi-infected mice. Front Immunol 2022; 13:1061031. [PMID: 36618364 PMCID: PMC9813216 DOI: 10.3389/fimmu.2022.1061031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Scrub typhus is a life-threatening zoonosis caused by the obligate intracellular bacterium Orientia tsutsugamushi (Ot) that is transmitted by the infected larvae of trombiculid mites. However, the mechanism by which Ot disseminates from the bite site to visceral organs remains unclear; host innate immunity against bacterial dissemination and replication during early infection is poorly understood. In this study, by using an intradermal infection mouse model and fluorescent probe-labeled Ot, we assessed the dynamic pattern of innate immune cell responses at the inoculation site. We found that neutrophils were the first responders to Ot infection and migrated into the skin for bacterial uptake. Ot infection greatly induced neutrophil activation, and Ot-neutrophil interaction remarkably promoted cell death both in vitro and in vivo. Depletion of neutrophils did not alter bacterial dissemination in mice, as evidenced by similar bacterial burdens in the skin and draining lymph nodes (dLN) at day 3, as well as in the lungs and brains at day 14, as compared to the control mice. Instead, dendritic cells (DCs) and macrophages played a role as a Trojan horse and transmitted Ot from the skin into dLN. Importantly, the absence of homing receptor CCR7 or neutralization of its ligand, CCL21, significantly impaired DC migration, resulting in reduced bacterial burdens in dLN. Taken together, our study sheds light on a CCR7/dendritic cell-mediated mechanism of early Ot dissemination and provides new insights into therapeutic and vaccine development strategies for scrub typhus.
Collapse
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| | - Hui Wang
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Casey Gonzales
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Joseph Thiriot
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Piyanate Sunyakumthorn
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Peter C. Melby
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
27
|
He Z, Wang C, Wang J, Zheng K, Ding N, Yu M, Li W, Tang Y, Li Y, Xiao J, Liang M, Wu Y. Chlamydia psittaci inhibits apoptosis of human neutrophils by activating P2X7 receptor expression. Int J Med Microbiol 2022; 312:151571. [PMID: 36511277 DOI: 10.1016/j.ijmm.2022.151571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/10/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
This study tested the hypothesis that Chlamydia psittaci (C. psittaci) survives and multiplies in human neutrophils by activating P2X7, a nonselective cationic channel receptor expressed constitutively on the surface of these cells. Findings illustrated that P2X7 receptor expression was enhanced in C. psittaci-infected neutrophils. C. psittaci was able to inhibite spontaneous apoptosis of neutrophils through mitochondrial-induced ATP release and IL-8 production. Importantly, inhibiting ATP activation of the P2X7 receptor with AZ10606120 promotes apoptosis, while stimulating P2X7 receptor expression with BzATP delayed spontaneous apoptosis of human neutrophils, suggesting that C. psittaci inhibits apoptosis of human neutrophils by activating P2X7 receptor. This study reveals new insights into the survival advantages of the latent persistent state of C. psittaci and the mechanism by which it evades the innate immune response.
Collapse
Affiliation(s)
- Zhangping He
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Chuan Wang
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Jianye Wang
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Kang Zheng
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Nan Ding
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Maoying Yu
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Weiwei Li
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Yuanyuan Tang
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Yumeng Li
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China
| | - Jian Xiao
- The Affiliated Nanhua Hospital, Department of laboratory medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Mingxing Liang
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
28
|
Clos J, Grünebast J, Holm M. Promastigote-to-Amastigote Conversion in Leishmania spp.-A Molecular View. Pathogens 2022; 11:1052. [PMID: 36145483 PMCID: PMC9503511 DOI: 10.3390/pathogens11091052] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
A key factor in the successful infection of a mammalian host by Leishmania parasites is their conversion from extracellular motile promastigotes into intracellular amastigotes. We discuss the physical and chemical triggers that induce this conversion and the accompanying changes at the molecular level crucial for the survival of these intracellular parasites. Special emphasis is given to the reliance of these trypanosomatids on the post-transcriptional regulation of gene expression but also to the role played by protein kinases, chaperone proteins and proteolytic enzymes. Lastly, we offer a model to integrate the transduction of different stress signals for the induction of stage conversion.
Collapse
|
29
|
Gurjar D, Kumar Patra S, Bodhale N, Lenka N, Saha B. Leishmania intercepts IFN-γR signaling at multiple levels in macrophages. Cytokine 2022; 157:155956. [PMID: 35785668 DOI: 10.1016/j.cyto.2022.155956] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022]
Abstract
IFN-γ, a type 2 interferon and a cytokine, is critical for both innate and adaptive immunity. IFN-γ binds to the IFN-γRs on the cell membrane of macrophages, signals through JAK1-STAT-1 pathway and induces IFN-γ-stimulated genes (ISGs). As Leishmania amastigotes reside and replicate within macrophages, IFN-γ mediated macrophage activation eventuate in Leishmania elimination. As befits the principle of parasitism, the impaired IFN-γ responsiveness in macrophages ensures Leishmania survival. IFN-γ responsiveness is a function of integrated molecular events at multiple levels in the cells that express IFN-γ receptors. In Leishmania-infected macrophages, reduced IFN-γRα expression, impaired IFN-γRα and IFN-γRβ hetero-dimerization due to altered membrane lipid composition, reduced JAK-1 and STAT-1 phosphorylation but increased STAT-1 degradation and impaired ISGs induction collectively determine the IFN-γ responsiveness and the efficacy of IFN-γ induced antileishmanial function of macrophages. Therefore, parasite load is not only decided by the levels of IFN-γ produced but also by the IFN-γ responsiveness. Indeed, in Leishmania-infected patients, IFN-γ is produced but IFN-γ signalling is downregulated. However, the molecular mechanisms of IFN-γ responsiveness remain unclear. Therefore, we review the current understanding of IFN-γ responsiveness of Leishmania-infected macrophages.
Collapse
Affiliation(s)
- Dhiraj Gurjar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | | | - Neelam Bodhale
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Nibedita Lenka
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
30
|
Venugopal G, Bird JT, Washam CL, Roys H, Bowlin A, Byrum SD, Weinkopff T. In vivo transcriptional analysis of mice infected with Leishmania major unveils cellular heterogeneity and altered transcriptomic profiling at single-cell resolution. PLoS Negl Trop Dis 2022; 16:e0010518. [PMID: 35789215 PMCID: PMC9286232 DOI: 10.1371/journal.pntd.0010518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/15/2022] [Accepted: 05/18/2022] [Indexed: 01/02/2023] Open
Abstract
Leishmania parasites cause cutaneous leishmaniasis (CL), a disease characterized by disfiguring, ulcerative skin lesions. Both parasite and host gene expression following infection with various Leishmania species has been investigated in vitro, but global transcriptional analysis following L. major infection in vivo is lacking. Thus, we conducted a comprehensive transcriptomic profiling study combining bulk RNA sequencing (RNA-Seq) and single-cell RNA sequencing (scRNA-Seq) to identify global changes in gene expression in vivo following L. major infection. Bulk RNA-Seq analysis revealed that host immune response pathways like the antigen processing and presentation pathway were significantly enriched amongst differentially expressed genes (DEGs) upon infection, while ribosomal pathways were significantly downregulated in infected mice compared to naive controls. scRNA-Seq analyses revealed cellular heterogeneity including distinct resident and recruited cell types in the skin following murine L. major infection. Within the individual immune cell types, several DEGs indicative of many interferon induced GTPases and antigen presentation molecules were significantly enhanced in the infected ears including macrophages, resident macrophages, and inflammatory monocytes. Ingenuity Pathway Analysis of scRNA-Seq data indicated the antigen presentation pathway was increased with infection, while EIF2 signaling is the top downregulated pathway followed by eIF4/p70S6k and mTOR signaling in multiple cell types including macrophages, blood and lymphatic endothelial cells. Altogether, this transcriptomic profile highlights known recruitment of myeloid cells to lesions and recognizes a potential role for EIF2 signaling in murine L. major infection in vivo.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jordan T. Bird
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Anne Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Arkansas Children’s Research Institute, Little Rock, Arkansas, United States of America
- * E-mail: (SDB); (TW)
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail: (SDB); (TW)
| |
Collapse
|
31
|
Nerb B, Dudziak D, Gessner A, Feuerer M, Ritter U. Have We Ignored Vector-Associated Microbiota While Characterizing the Function of Langerhans Cells in Experimental Cutaneous Leishmaniasis? FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.874081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
32
|
Pellegrini JM, Gorvel JP, Mémet S. Immunosuppressive Mechanisms in Brucellosis in Light of Chronic Bacterial Diseases. Microorganisms 2022; 10:1260. [PMID: 35888979 PMCID: PMC9324529 DOI: 10.3390/microorganisms10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Brucellosis is considered one of the major zoonoses worldwide, constituting a critical livestock and human health concern with a huge socio-economic burden. Brucella genus, its etiologic agent, is composed of intracellular bacteria that have evolved a prodigious ability to elude and shape host immunity to establish chronic infection. Brucella's intracellular lifestyle and pathogen-associated molecular patterns, such as its specific lipopolysaccharide (LPS), are key factors for hiding and hampering recognition by the immune system. Here, we will review the current knowledge of evading and immunosuppressive mechanisms elicited by Brucella species to persist stealthily in their hosts, such as those triggered by their LPS and cyclic β-1,2-d-glucan or involved in neutrophil and monocyte avoidance, antigen presentation impairment, the modulation of T cell responses and immunometabolism. Attractive strategies exploited by other successful chronic pathogenic bacteria, including Mycobacteria, Salmonella, and Chlamydia, will be also discussed, with a special emphasis on the mechanisms operating in brucellosis, such as granuloma formation, pyroptosis, and manipulation of type I and III IFNs, B cells, innate lymphoid cells, and host lipids. A better understanding of these stratagems is essential to fighting bacterial chronic infections and designing innovative treatments and vaccines.
Collapse
|
33
|
Quintela-Carvalho G, Goicochea AMC, Mançur-Santos V, Viana SDM, Luz YDS, Dias BRS, Lázaro-Souza M, Suarez M, de Oliveira CI, Saraiva EM, Brodskyn CI, Veras PT, de Menezes JP, Andrade BB, Lima JB, Descoteaux A, Borges VM. Leishmania infantum Defective in Lipophosphoglycan Biosynthesis Interferes With Activation of Human Neutrophils. Front Cell Infect Microbiol 2022; 12:788196. [PMID: 35463648 PMCID: PMC9019130 DOI: 10.3389/fcimb.2022.788196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Visceral leishmaniasis (VL) is often associated with hematologic manifestations that may interfere with neutrophil response. Lipophosphoglycan (LPG) is a major molecule on the surface of Leishmania promastigotes, which has been associated with several aspects of the parasite–vector–host interplay. Here, we investigated how LPG from Leishmania (L.) infantum, the principal etiological agent of VL in the New World, influences the initial establishment of infection during interaction with human neutrophils in an experimental setting in vitro. Human neutrophils obtained from peripheral blood samples were infected with either the wild-type L. infantum (WT) strain or LPG-deficient mutant (∆lpg1). In this setting, ∆lpg1 parasites displayed reduced viability compared to WT L. infantum; such finding was reverted in the complemented ∆lpg1+LPG1 parasites at 3- and 6-h post-infection. Confocal microscopy experiments indicated that this decreased survival was related to enhanced lysosomal fusion. In fact, LPG-deficient L. infantum parasites more frequently died inside neutrophil acidic compartments, a phenomenon that was reverted when host cells were treated with Wortmannin. We also observed an increase in the secretion of the neutrophil collagenase matrix metalloproteinase-8 (MMP-8) by cells infected with ∆lpg1 L. infantum compared to those that were infected with WT parasites. Furthermore, collagen I matrix degradation was found to be significantly increased in ∆lpg1 parasite-infected cells but not in WT-infected controls. Flow cytometry analysis revealed a substantial boost in production of reactive oxygen species (ROS) during infection with either WT or ∆lpg1 L. infantum. In addition, killing of ∆lpg1 parasites was shown to be more dependent on the ROS production than that of WT L. infantum. Notably, inhibition of the oxidative stress with Apocynin potentially fueled ∆lpg1 L. infantum fitness as it increased the intracellular parasite viability. Thus, our observations demonstrate that LPG may be a critical molecule fostering parasite survival in human neutrophils through a mechanism that involves cellular activation and generation of free radicals.
Collapse
Affiliation(s)
- Graziele Quintela-Carvalho
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia Baiano (IFBaiano), Alagoinhas, Brazil
| | - Astrid Madeleine Calero Goicochea
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Vanessa Mançur-Santos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Sayonara de Melo Viana
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Yasmin da Silva Luz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Beatriz Rocha Simões Dias
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Milena Lázaro-Souza
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Martha Suarez
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Camila Indiani de Oliveira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Elvira M. Saraiva
- Departamento de Imunologia, Laboratório de Imunobiologia das Leishmanioses, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudia I. Brodskyn
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Patrícia T. Veras
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Juliana P.B. de Menezes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Bruno B. Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil
- Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
| | - Jonilson Berlink Lima
- Núcleo de Agentes Infecciosos e Vetores (NAIVE), Universidade Federal do Oeste da Bahia (UFOB), Barreiras, Brazil
| | - Albert Descoteaux
- Institut National de la Recherche Scientifique (INRS)–Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
- *Correspondence: Valéria M. Borges, ; Albert Descoteaux,
| | - Valéria M. Borges
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
- *Correspondence: Valéria M. Borges, ; Albert Descoteaux,
| |
Collapse
|
34
|
Immune Responses in Leishmaniases: An Overview. Trop Med Infect Dis 2022; 7:tropicalmed7040054. [PMID: 35448829 PMCID: PMC9029249 DOI: 10.3390/tropicalmed7040054] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a parasitic, widespread, and neglected disease that affects more than 90 countries in the world. More than 20 Leishmania species cause different forms of leishmaniasis that range in severity from cutaneous lesions to systemic infection. The diversity of leishmaniasis forms is due to the species of parasite, vector, environmental and social factors, genetic background, nutritional status, as well as immunocompetence of the host. Here, we discuss the role of the immune system, its molecules, and responses in the establishment, development, and outcome of Leishmaniasis, focusing on innate immune cells and Leishmania major interactions.
Collapse
|
35
|
Magold AI, Swartz MA. Pathogenic Exploitation of Lymphatic Vessels. Cells 2022; 11:979. [PMID: 35326430 PMCID: PMC8946894 DOI: 10.3390/cells11060979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Lymphatic vessels provide a critical line of communication between peripheral tissues and their draining lymph nodes, which is necessary for robust immune responses against infectious agents. At the same time, lymphatics help shape the nature and kinetics of immune responses to ensure resolution, limit tissue damage, and prevent autoimmune responses. A variety of pathogens have developed strategies to exploit these functions, from multicellular organisms like nematodes to bacteria, viruses, and prions. While lymphatic vessels serve as transport routes for the dissemination of many pathogens, their hypoxic and immune-suppressive environments can provide survival niches for others. Lymphatics can be exploited as perineural niches, for inter-organ distribution among highly motile carrier cells, as effective replicative niches, and as alternative routes in response to therapy. Recent studies have broadened our understanding of lymphatic involvement in pathogenic spread to include a wider range of pathogens, as well as new mechanisms of exploitation, which we summarize here.
Collapse
Affiliation(s)
- Alexandra I. Magold
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA;
| | - Melody A. Swartz
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA;
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
36
|
Taslimi Y, Masoudzadeh N, Bahrami F, Rafati S. Cutaneous leishmaniasis: multiomics approaches to unravel the role of immune cells checkpoints. Expert Rev Proteomics 2022; 19:213-225. [PMID: 36191333 DOI: 10.1080/14789450.2022.2131545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Cutaneous leishmaniasis (CL) is the most frequent form of leishmaniases, associated with skin inflammation and ulceration. Understanding the interaction of different phagocytic cells in the recognition and uptake of different Leishmania species is critical for controlling the infection. Phagocytic cells have a pivotal role as professional antigen-presenting cells that bridge the innate and adaptive immunity and shape the outcome of the disease. AREAS COVERED Here we reviewed new technologies with high-throughput data collection capabilities along with systems biology approaches which are recently being used to decode the paradox of CL immunology. EXPERT OPINION We emphasized on the crosstalk between DC and T-cells while focusing on the immune checkpoints interactions between the human immune system and the Leishmania species. Further, we discussed omics technologies including bulk RNA sequencing, reverse transcriptase-multiplex ligation dependent probe amplification (RT-MLPA), and proximity extension assay (PEA) in studies on human blood or tissue-driven samples from CL patients in which we have so far been involved.
Collapse
Affiliation(s)
- Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| |
Collapse
|
37
|
Chan A, Ayala JM, Alvarez F, Piccirillo C, Dong G, Langlais D, Olivier M. The role of Leishmania GP63 in the modulation of innate inflammatory response to Leishmania major infection. PLoS One 2022; 16:e0262158. [PMID: 34972189 PMCID: PMC8719666 DOI: 10.1371/journal.pone.0262158] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/16/2021] [Indexed: 01/08/2023] Open
Abstract
Leishmaniasis is a disease caused by the protozoan parasite Leishmania and is known to affect millions of individuals worldwide. In recent years, we have established the critical role played by Leishmania zinc-metalloprotease GP63 in the modulation of host macrophage signalling and functions, favouring its survival and progression within its host. Leishmania major lacking GP63 was reported to cause limited infection in mice, however, it is still unclear how GP63 may influence the innate inflammatory response and parasite survival in an in vivo context. Therefore, we were interested in analyzing the early innate inflammatory events upon Leishmania inoculation within mice and establish whether Leishmania GP63 influences this initial inflammatory response. Experimentally, L. major WT (L. majorWT), L. major GP63 knockout (L. majorKO), or L. major GP63 rescue (L. majorR) were intraperitoneally inoculated in mice and the inflammatory cells recruited were characterized microscopically and by flow cytometry (number and cell type), and their infection determined. Pro-inflammatory markers such as cytokines, chemokines, and extracellular vesicles (EVs, e.g. exosomes) were monitored and proteomic analysis was performed on exosome contents. Data obtained from this study suggest that Leishmania GP63 does not significantly influence the pathogen-induced inflammatory cell recruitment, but rather their activation status and effector function. Concordantly, internalization of promastigotes during early infection could be influenced by GP63 as fewer L. majorKO amastigotes were found within host cells and appear to maintain in host cells over time. Collectively this study provides a clear analysis of innate inflammatory events occurring during L. major infection and further establish the prominent role of the virulence factor GP63 to provide favourable conditions for host cell infection.
Collapse
Affiliation(s)
- Aretha Chan
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Jose-Mauricio Ayala
- Department of Human Genetics, McGill Genome Centre, Montréal, QC, Canada
- McGill Research Centre on Complex Traits, Montreal, QC, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- FOCiS Centre of Excellence in Translational Immunology, Montréal, QC, Canada
| | - Ciriaco Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill Research Centre on Complex Traits, Montreal, QC, Canada
- FOCiS Centre of Excellence in Translational Immunology, Montréal, QC, Canada
| | - George Dong
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill Research Centre on Complex Traits, Montreal, QC, Canada
| | - David Langlais
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill Genome Centre, Montréal, QC, Canada
- McGill Research Centre on Complex Traits, Montreal, QC, Canada
- * E-mail: (MO); (DL)
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill Research Centre on Complex Traits, Montreal, QC, Canada
- FOCiS Centre of Excellence in Translational Immunology, Montréal, QC, Canada
- * E-mail: (MO); (DL)
| |
Collapse
|
38
|
Yu Y, Li J, Zhang Y, Ma Z, Sun H, Wei X, Bai Y, Wu Z, Zhang X. A bioinspired hierarchical nanoplatform targeting and responding to intracellular pathogens to eradicate parasitic infections. Biomaterials 2021; 280:121309. [PMID: 34896862 DOI: 10.1016/j.biomaterials.2021.121309] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/02/2022]
Abstract
Intracellular bacteria-mediated antibiotic tolerance, which acts as a "Trojan horse," plays a critical and underappreciated role in chronic and recurrent infections. Failure of conventional antibiotic therapy is often encountered because infected cells prevent drug permeation or the drug concentration is too low at the site of resident bacteria. New paradigms are therefore urgently needed for intracellular anti-infective therapy. Here, a novel therapeutic was developed for targeted delivery of antibiotics into bacteria-infected macrophages to improve drug accumulation in intracellular niches and bactericidal activity of antibiotics against intracellular pathogens. This hierarchical nanoplatform includes a glycocalyx-mimicking shell that enables rapid uptake by macrophages. Subsequently, the targeting moieties are activated in response to the bacteria, and the release of entrapped antibiotics is triggered by bacteria and bacteria-secreted enzymes. The self-immolative drug delivery nanoplatform eliminates intracellular pathogenic bacteria residing in macrophages more efficiently compared to drugs alone. The in vivo dynamically monitored nanosystem also efficiently inhibited the growth of intracellular Staphylococcus aureus in infected muscles of mice with negligible systemic toxicity. The novel dual-targeting design of an all-in-one therapeutic platform can be used as an alternative strategy to reanimate antibiotic therapy against multifarious intracellular bacterial infections.
Collapse
Affiliation(s)
- Yunjian Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Zhuang Ma
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Haonan Sun
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Xiaosong Wei
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Yayun Bai
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Zhongming Wu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
39
|
Gomes RS, Silva MVT, Oliveira MAP, Joosten LAB, Ribeiro-Dias F. Protective immune response mediated by neutrophils in experimental visceral leishmaniasis is enhanced by IL-32γ. Cell Immunol 2021; 371:104449. [PMID: 34784560 DOI: 10.1016/j.cellimm.2021.104449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/03/2022]
Abstract
Neutrophils are important cells in protection against microbial infections including visceral leishmaniasis (VL). It is well known that IL-32γ increases the protective T helper 17 cell mediated immune response against Leishmania infantum. Thus, in this study we evaluated whether IL-32 γ can increase the protective role of neutrophils against VL. In comparison with wild type (WT) mice, transgenic mice for human IL-32 γ (IL-32 γ Tg) presented a higher frequency and absolute number of neutrophils in both spleen and liver after the establishment of L. infantum infection. The IL-32 concentrations correlated with neutrophil numbers in the infected tissues. The IL-32 γ -induced recruitment of neutrophils was dependent on IL-17, since inhibition of Th17 T cells generation and IL-17 production with digoxin treatment reversed the effects of IL-32 γ. In murine neutrophils, the presence of IL-32 γ enhanced the phagocytosis of L. infantum via CR3. In addition, murine IL-32 γ Tg neutrophils were able to kill L. infantum due to the increased production of ROS when compared with WT neutrophils. In fact, IL-32 γ Tg mice lost their ability to control infection by L. infantum when neutrophils were depleted. In parallel, treatment of human neutrophils with recombinant IL-32 γ increased phagocytosis and ROS-dependent killing of L. infantum, similarly to murine IL-32 γ Tg neutrophils. The data show that IL-32 γ induces neutrophil recruitment to organs affected by VL and increases phagocytosis and killing of L. infantum by neutrophils. Together, data indicate the pivotal axis IL-32 γ -Th17-neutrophils to control VL.
Collapse
Affiliation(s)
- Rodrigo Saar Gomes
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil.
| | | | | | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil.
| |
Collapse
|
40
|
Chemokines in Leishmaniasis: Map of cell movements highlights the landscape of infection and pathogenesis. Cytokine 2021; 147:155339. [DOI: 10.1016/j.cyto.2020.155339] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
|
41
|
He Z, Xiao J, Wang J, Lu S, Zheng K, Yu M, Liu J, Wang C, Ding N, Liang M, Wu Y. The Chlamydia psittaci Inclusion Membrane Protein 0556 Inhibits Human Neutrophils Apoptosis Through PI3K/AKT and NF-κB Signaling Pathways. Front Immunol 2021; 12:694573. [PMID: 34484191 PMCID: PMC8414580 DOI: 10.3389/fimmu.2021.694573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/23/2021] [Indexed: 01/09/2023] Open
Abstract
Inclusion membrane proteins (Incs) play an important role in the structure and stability of chlamydial inclusion and the interaction between Chlamydia spp. and their hosts. Following Chlamydia infection through the respiratory tract, human polymorphonuclear neutrophils (hPMN) not only act as the primary immune cells reaching the lungs, but also serve as reservoir for Chlamydia. We have previously identified a Chlamydia psittaci hypothetical protein, CPSIT_0556, as a medium expressed inclusion membrane protein. However, the role of inclusion membrane protein, CPSIT_0556 in regulating hPMN functions remains unknown. In the present study, we found that CPSIT_0556 could not only inhibit hPMN apoptosis through the PI3K/Akt and NF-κB signaling pathways by releasing IL-8, but also delays procaspase-3 processing and inhibits caspase-3 activity in hPMN. Up-regulating the expression of anti-apoptotic protein Mcl-1 and down-regulating the expression of pro-apoptotic protein Bax could also inhibit the translocalization of Bax in the cytoplasm into the mitochondria, as well as induce the transfer of p65 NF-κB from the cytoplasm to the nucleus. Overall, our findings demonstrate that CPSIT_0556 could inhibit hPMN apoptosis through PI3K/Akt and NF-κB pathways and provide new insights towards understanding a better understanding of the molecular pathogenesis and immune escape mechanisms of C. psittaci.
Collapse
Affiliation(s)
- Zhangping He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Jian Xiao
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital of University of South China, Hengyang, China
| | - Jianye Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Simin Lu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Kang Zheng
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Maoying Yu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Jie Liu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Chuan Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Nan Ding
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| | - Mingxing Liang
- Department of Clinical Laboratory, The Affiliated Huaihua Hospital of University of South China, Huaihua, China
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, China
| |
Collapse
|
42
|
Chanyalew M, Abebe M, Endale B, Girma S, Tasew G, Bobosha K, Zewide M, Howe R, van Zandbergen G, Ritter U, Gadisa E, Aseffa A, Laskay T. Enhanced activation of blood neutrophils and monocytes in patients with Ethiopian localized cutaneous leishmaniasis in response to Leishmania aethiopica Neutrophil activation in Ethiopian cutaneous leishmaniasis. Acta Trop 2021; 220:105967. [PMID: 34029532 DOI: 10.1016/j.actatropica.2021.105967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/04/2021] [Accepted: 05/16/2021] [Indexed: 11/15/2022]
Abstract
Recent studies suggest an essential role of the innate immune effector cells neutrophils and monocytes in protection or disease progression in the early course of Leishmania infection. In areas endemic for cutaneous leishmaniasis in Ethiopia most individuals are exposed to bites of infected sandflies. Still only a minor ratio of the inhabitants develops symptomatic disease. Neutrophils, followed by monocytes, are the first cells to be recruited to the site of Leishmania infection, the initial response of neutrophils to parasites appears to be crucial for the protective response and disease outcome. Our working hypothesis is that neutrophils and/or monocytes in localized cutaneous leishmaniasis (LCL) patients may have defects in function of innate immune cell that contribute to failure to parasite clearance that lead to establishment of infection. The response of cells in Ethiopian LCL patients and healthy controls to Leishmania aethiopica and to the Toll like receptor (TLR) agonists lipopolysaccharide (LPS) and macrophage activating lipopeptide-2 (MALP-2) was investigated by assessing the cell surface expression of CD62L (on neutrophil and monocyte) and CD66b (only on neutrophil), as well as reactive oxygen species (ROS) production by using whole blood-based assays in vitro. No impaired response of neutrophils and monocytes to the microbial constituents LPS and MALP-2 was observed. Neutrophils and monocytes from LCL patients responded stronger to Leishmania aethiopica in the applied whole blood assays than cells from healthy individuals. These experimental findings do not support the hypothesis regarding a possible dysfunction of neutrophils and monocytes in cutaneous leishmaniasis. On the contrary, these cells react stronger in LCL patients as compared to healthy controls. The differential response to L. aethiopica observed between LCL patients and healthy controls have the potential to serve as biomarker to develop FACS based diagnostic/ prognostic techniques for LCL.
Collapse
Affiliation(s)
- Menberework Chanyalew
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia.
| | - Markos Abebe
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Birtukan Endale
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Selfu Girma
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Geremew Tasew
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia; Leishmaniasis Research Laboratory, Ethiopian Public Health Institute, P.O. Box 1242, Addis Ababa, Ethiopia
| | - Kidist Bobosha
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Martha Zewide
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia.
| | - Rawleigh Howe
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Ger van Zandbergen
- Division of Immunology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen D-63225, Germany.
| | - Uwe Ritter
- Regensburg Center for Interventional Immunology (RCI), Institute of Immunology, University Medical Center Regensburg, University of Regensburg, Regensburg D-93053, Germany.
| | - Endalamaw Gadisa
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Abraham Aseffa
- Research and Innovation Division, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Tamás Laskay
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck D-23560, Germany.
| |
Collapse
|
43
|
Sandfly Fever Sicilian Virus-Leishmania major co-infection modulates innate inflammatory response favoring myeloid cell infections and skin hyperinflammation. PLoS Negl Trop Dis 2021; 15:e0009638. [PMID: 34310619 PMCID: PMC8341699 DOI: 10.1371/journal.pntd.0009638] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/05/2021] [Accepted: 07/09/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The leishmaniases are a group of sandfly-transmitted diseases caused by species of the protozoan parasite, Leishmania. With an annual incidence of 1 million cases, 1 billion people living in Leishmania-endemic regions, and nearly 30,000 deaths each year, leishmaniasis is a major global public health concern. While phlebotomine sandflies are well-known as vectors of Leishmania, they are also the vectors of various phleboviruses, including Sandfly Fever Sicilian Virus (SFSV). Cutaneous leishmaniasis (CL), caused by Leishmania major (L. major), among other species, results in development of skin lesions on the infected host. Importantly, there exists much variation in the clinical manifestation between individuals. We propose that phleboviruses, vectored by and found in the same sandfly guts as Leishmania, may be a factor in determining CL severity. It was reported by our group that Leishmania exosomes are released into the gut of the sandfly vector and co-inoculated during blood meals, where they exacerbate CL skin lesions. We hypothesized that, when taking a blood meal, the sandfly vector infects the host with Leishmania parasites and exosomes as well as phleboviruses, and that this viral co-infection results in a modulation of leishmaniasis. METHODOLOGY/PRINCIPAL FINDINGS In vitro, we observed modulation by SFSV in MAP kinase signaling as well as in the IRF3 pathway that resulted in a pro-inflammatory phenotype. Additionally, we found that SFSV and L. major co-infection resulted in an exacerbation of leishmaniasis in vivo, and by using endosomal (Toll-like receptor) TLR3, and MAVS knock-out mice, deduced that SFSV's hyperinflammatory effect was TLR3- and MAVS-dependent. Critically, we observed that L. major and SFSV co-infected C57BL/6 mice demonstrated significantly higher parasite burden than mice solely infected with L. major. Furthermore, viral presence increased leukocyte influx in vivo. This influx was accompanied by elevated total extracellular vesicle numbers. Interestingly, L. major displayed higher infectiveness with coincident phleboviral infection compared to L. major infection alone. CONCLUSION/SIGNIFICANCE Overall our work represents novel findings that contribute towards understanding the causal mechanisms governing cutaneous leishmaniasis pathology. Better comprehension of the potential role of viral co-infection could lead to treatment regimens with enhanced effectiveness.
Collapse
|
44
|
Ly6G deficiency alters the dynamics of neutrophil recruitment and pathogen capture during Leishmania major skin infection. Sci Rep 2021; 11:15071. [PMID: 34302006 PMCID: PMC8302578 DOI: 10.1038/s41598-021-94425-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/05/2021] [Indexed: 01/21/2023] Open
Abstract
Neutrophils represent one of the first immune cell types recruited to sites of infection, where they can control pathogens by phagocytosis and cytotoxic mechanisms. Intracellular pathogens such as Leishmania major can hijack neutrophils to establish an efficient infection. However the dynamic interactions of neutrophils with the pathogen and other cells at the site of the infection are incompletely understood. Here, we have investigated the role of Ly6G, a homolog of the human CD177 protein, which has been shown to interact with cell adhesion molecules, and serves as a bona fide marker for neutrophils in mice. We show that Ly6G deficiency decreases the initial infection rate of neutrophils recruited to the site of infection. Although the uptake of L. major by subsequently recruited monocytes was tightly linked with the concomitant uptake of neutrophil material, this process was not altered by Ly6G deficiency of the neutrophils. Instead, we observed by intravital 2-photon microscopy that Ly6G-deficient neutrophils entered the site of infection with delayed initial recruitment kinetics. Thus, we conclude that by promoting neutrophils’ ability to efficiently enter the site of infection, Ly6G contributes to the early engagement of intracellular pathogens by the immune system.
Collapse
|
45
|
de Sousa Gonçalves R, de Pinho FA, Dinis-Oliveira RJ, Mendes MO, de Andrade TS, da Silva Solcà M, Larangeira DF, Silvestre R, Barrouin-Melo SM. Nutritional adjuvants with antioxidant properties in the treatment of canine leishmaniasis. Vet Parasitol 2021; 298:109526. [PMID: 34271314 DOI: 10.1016/j.vetpar.2021.109526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/14/2021] [Accepted: 07/05/2021] [Indexed: 01/03/2023]
Abstract
Clinical improvement of dogs treated for canine leishmaniasis (CanL) requires reducing Leishmania infantum loads, which depend on intracellular oxidant compounds to destroy the parasite. However, oxidative species' excess and antioxidants consumption can culminate in oxidative stress, resulting in increased, widespread inflammation. We aimed to evaluate if early or late addition of nutritional adjuvants (NAs) - omega-3 polyunsaturated fatty acids and B vitamins - to anti-Leishmania drugs (ALDs) in the treatment of CanL would be clinically beneficial. For that, serum biomarkers including oxidative stress parameters were analyzed during 12 months in dogs allocated to two treatment groups: (G1) NAs administered from 30 days prior to the beginning of ALDs; and (G2) NAs administered from 61 days after the beginning of ALDs. Both G1 and G2 continued to receive NAs until the 12th month. The ALDs administered were metronidazole associated with ketoconazole (40 days), followed by allopurinol from day 41 until the 12th month. G1 exhibited superior inflammation control, with reduced globulins (p = 0.025), specific anti-Leishmania immunoglobulins (p = 0.016), total protein (p = 0.031), and an increased serum albumin/globulin ratio (p = 0.033), compared to G2. The early use of NAs associated with ALDs is clinically beneficial in treating dogs with CanL.
Collapse
Affiliation(s)
- Rafaela de Sousa Gonçalves
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia, 40170-110, Salvador, BA, Brazil; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
| | - Flaviane Alves de Pinho
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia, 40170-110, Salvador, BA, Brazil; Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, 40170-110, Salvador, BA, Brazil
| | - Ricardo Jorge Dinis-Oliveira
- TOXRUN - Toxicology Research Unit, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, Rua Central de Gandra, 1317, 4585-116, Gandra, Portugal; Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal; UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Mariana Oliveira Mendes
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia, 40170-110, Salvador, BA, Brazil
| | - Tiago Sena de Andrade
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia, 40170-110, Salvador, BA, Brazil
| | - Manuela da Silva Solcà
- Department of Preventive Veterinary Medicine and Animal Production of the School of Veterinary Medicine and Zootechny, UFBA, 40170-110, Salvador, Bahia, Brazil
| | - Daniela Farias Larangeira
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia, 40170-110, Salvador, BA, Brazil; Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, 40170-110, Salvador, BA, Brazil
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal; ICVS/3B's Associate Laboratory, 4710-057, Braga, Portugal
| | - Stella Maria Barrouin-Melo
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia, 40170-110, Salvador, BA, Brazil; Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, 40170-110, Salvador, BA, Brazil.
| |
Collapse
|
46
|
Guimaraes-Costa AB, Shannon JP, Waclawiak I, Oliveira J, Meneses C, de Castro W, Wen X, Brzostowski J, Serafim TD, Andersen JF, Hickman HD, Kamhawi S, Valenzuela JG, Oliveira F. A sand fly salivary protein acts as a neutrophil chemoattractant. Nat Commun 2021; 12:3213. [PMID: 34050141 PMCID: PMC8163758 DOI: 10.1038/s41467-021-23002-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/09/2021] [Indexed: 01/10/2023] Open
Abstract
Apart from bacterial formyl peptides or viral chemokine mimicry, a non-vertebrate or insect protein that directly attracts mammalian innate cells such as neutrophils has not been molecularly characterized. Here, we show that members of sand fly yellow salivary proteins induce in vitro chemotaxis of mouse, canine and human neutrophils in transwell migration or EZ-TAXIScan assays. We demonstrate murine neutrophil recruitment in vivo using flow cytometry and two-photon intravital microscopy in Lysozyme-M-eGFP transgenic mice. We establish that the structure of this ~ 45 kDa neutrophil chemotactic protein does not resemble that of known chemokines. This chemoattractant acts through a G-protein-coupled receptor and is dependent on calcium influx. Of significance, this chemoattractant protein enhances lesion pathology (P < 0.0001) and increases parasite burden (P < 0.001) in mice upon co-injection with Leishmania parasites, underlining the impact of the sand fly salivary yellow proteins on disease outcome. These findings show that some arthropod vector-derived factors, such as this chemotactic salivary protein, activate rather than inhibit the host innate immune response, and that pathogens take advantage of these inflammatory responses to establish in the host. Immune mimicry has been shown in chemokine like moieties from bacteria and viruses. Here, the authors characterise a sand fly salivary protein that induces neutrophil chemotaxis and explore its impact in a model of parasitic infection.
Collapse
Affiliation(s)
- Anderson B Guimaraes-Costa
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.,Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - John P Shannon
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ingrid Waclawiak
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Jullyanna Oliveira
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Waldione de Castro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Xi Wen
- Chemotaxis Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Joseph Brzostowski
- Twinbrook Imaging Facility, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, USA
| | - Tiago D Serafim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - John F Andersen
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
47
|
Maksouri H, Darif D, Estaquier J, Riyad M, Desterke C, Lemrani M, Dang PMC, Akarid K. The Modulation of NADPH Oxidase Activity in Human Neutrophils by Moroccan Strains of Leishmania major and Leishmania tropica Is Not Associated with p47 phox Phosphorylation. Microorganisms 2021; 9:1025. [PMID: 34068760 PMCID: PMC8151549 DOI: 10.3390/microorganisms9051025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 11/30/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the first phagocyte recruited and infected by Leishmania. They synthetize superoxide anions (O2-) under the control of the NADPH oxidase complex. In Morocco, Leishmania major and L. tropica are the main species responsible for cutaneous leishmaniasis (CL). The impact of these parasites on human PMN functions is still unclear. We evaluated the in vitro capacity of primary Moroccan strains of L. major and L. tropica to modulate PMN O2- production and p47phox phosphorylation status of the NADPH oxidase complex. PMNs were isolated from healthy blood donors, and their infection rate was measured by microscopy. O2- production was measured by superoxide dismutase-inhibitable reduction of cytochrome C. P47phox phosphorylation was analyzed by Western blot using specific antibodies against Ser328 and Ser345 sites. Whereas we did not observe any difference in PMN infectivity rate, our results indicated that only L. tropica promastigotes inhibited both fMLF- and PMA-mediated O2- production independently of p47phox phosphorylation. Leishmania soluble antigens (SLAs) from both species significantly inhibited O2- induced by fMLF or PMA. However, they only decreased PMA-induced p47phox phosphorylation. L. major and L. tropica modulated differently O2- production by human PMNs independently of p47phox phosphorylation. The inhibition of ROS production by L. tropica could be a mechanism of its survival within PMNs that might explain the reported chronic pathogenicity of L. tropica CL.
Collapse
Affiliation(s)
- Hasnaa Maksouri
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy (FMPC), Hassan II University of Casablanca (UH2C), 20000 Casablanca, Morocco; (H.M.); (M.R.)
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| | - Dounia Darif
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| | - Jerome Estaquier
- INSERM U1124, Paris University, 75006 Paris, France
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medicine, Laval University, Quebec City, QC G1V0A6, Canada
| | - Myriam Riyad
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy (FMPC), Hassan II University of Casablanca (UH2C), 20000 Casablanca, Morocco; (H.M.); (M.R.)
| | - Christophe Desterke
- Faculty of Medicine of the Kremlin-Bicêtre, University Paris-Sud, 94270 Paris, France;
| | - Meryem Lemrani
- Laboratory of Parasitology and Vector-Borne-Diseases, Institut Pasteur du Maroc, 20250 Casablanca, Morocco;
| | - Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, 75018 Paris, France;
- Inflamex Laboratory of Excellence, Faculty of Medicine, Site Xavier Bichat, University of Paris, 75018 Paris, France
| | - Khadija Akarid
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| |
Collapse
|
48
|
Jobin K, Müller DN, Jantsch J, Kurts C. Sodium and its manifold impact on our immune system. Trends Immunol 2021; 42:469-479. [PMID: 33962888 DOI: 10.1016/j.it.2021.04.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022]
Abstract
The Western diet is rich in salt, and a high salt diet (HSD) is suspected to be a risk factor for cardiovascular diseases. It is now widely accepted that an experimental HSD can stimulate components of the immune system, potentially exacerbating certain autoimmune diseases, or alternatively, improving defenses against certain infections, such as cutaneous leishmaniasis. However, recent findings show that an experimental HSD may also aggravate other infections (e.g., pyelonephritis or systemic listeriosis). Here, we discuss the modulatory effects of a HSD on the microbiota, metabolic signaling, hormonal responses, local sodium concentrations, and their effects on various immune cell types in different tissues. We describe how these factors are integrated, resulting either in immune stimulation or suppression in various tissues and disease settings.
Collapse
Affiliation(s)
- Katarzyna Jobin
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany; Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, Würzburg, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, and Max Delbruck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg and University of Regensburg, Regensburg, Germany.
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia.
| |
Collapse
|
49
|
Pugliese M, Sfacteria A, Oliva G, Falcone A, Gizzarelli M, Passantino A. Clinical Significance of ROMs, OXY, SHp and HMGB-1 in Canine Leishmaniosis. Animals (Basel) 2021; 11:754. [PMID: 33803468 PMCID: PMC7998487 DOI: 10.3390/ani11030754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate the role of oxidative stress parameters (ROMs, OXY, SHp), the Oxidative Stress index (OSi), and High Mobility Group Box-1 protein (HMGB-1) in canine leishmaniosis (CanL). For this study, thirty dogs, naturally infected with Leishmania spp. (Leishmania Group, LEISH) and ten healthy adult dogs (control group, CTR) were included. The diagnosis of CanL was performed by a cytological examination of lymph nodes, real time polymerase chain reaction on biological tissues (lymph nodes and whole blood), and an immunofluorescence antibody test (IFAT) for the detection of anti-Leishmania antibodies associated with clinical signs such as dermatitis, lymphadenopathy, onychogryphosis, weight loss, cachexia, lameness, conjunctivitis, epistaxis, and hepatosplenomegaly. The HMGB-1 and oxidative stress parameters of the LEISH Group were compared with the values recorded in the CTR group (Mann Whitney Test, p < 0.05). Spearman rank correlation was applied to evaluate the correlation between the HMGB-1, oxidative stress biomarkers, hematological and biochemical parameters in the LEISH Group. Results showed statistically significant higher values of SHp in the LEISH Group. Specific correlation between the ROMs and the number of red blood cells, and between HGMB-1 and SHp were recorded. These preliminary data may suggest the potential role of oxidative stress in the pathogenesis of CanL. Further studies are undoubtedly required to evaluate the direct correlation between inflammation parameters with the different stages of CanL. Similarly, further research should investigate the role of ROMs in the onset of anemia.
Collapse
Affiliation(s)
- Michela Pugliese
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.P.); (A.S.); (A.F.); (A.P.)
| | - Alessandra Sfacteria
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.P.); (A.S.); (A.F.); (A.P.)
| | - Gaetano Oliva
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy;
| | - Annastella Falcone
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.P.); (A.S.); (A.F.); (A.P.)
| | - Manuela Gizzarelli
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy;
| | - Annamaria Passantino
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.P.); (A.S.); (A.F.); (A.P.)
| |
Collapse
|
50
|
Redox Status in Canine Leishmaniasis. Animals (Basel) 2021; 11:ani11010119. [PMID: 33429894 PMCID: PMC7828002 DOI: 10.3390/ani11010119] [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: 12/01/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Leishmaniasis is under strict observation by World Health Organization but its pathogenesis has not been completely clarified yet. Our aim was to compare healthy and affected dogs measuring parameters related to oxidative stress, namely reactive oxygen species, reactive nitrogen species and scavenger activities, using colorimetric assays. Our results demonstrate that several of the examined parameters are modified in canine Leishmaniasis. Therefore, it is essential to further investigate this topic to shed light on the pathogenesis of the disease. Abstract The World Health Organization defined leishmaniasis as one of the priority attention diseases. Aiming to clarify some aspects of its pathogenetic mechanisms, our study focused on the assessment of redox status in dogs, the main reservoir for Leishmania infantum. Forty-five dogs from an endemic area in southern Italy were divided into four different groups (from mild disease with negative to low positive antibody levels to very severe disease with medium to high positive antibody levels) according to the LeishVet group guidelines. Their plasma and/or sera were tested for reactive oxygen species (ROS), namely the superoxide anion (O2−), reactive nitrogen species (RNS), such as nitric oxide (NO) and hydroperoxides (ROOH), as well as activity of the detoxifying enzyme superoxide dismutase (SOD), and total nonenzymatic antioxidant capacity, as determined by the ferric reducing-antioxidant power (FRAP) assay. O2− generation was significantly (p < 0.05) reduced in leishmaniasis-affected dogs independently of the clinical stage, while NO production was stimulated (p < 0.05) only in II and III stage patients. No difference could be found for the levels of hydroperoxides and SOD activity between healthy and pathological subjects. FRAP values were lower in affected dogs but only in stage II. Taken together, although we demonstrated that several redox status parameters are altered in the plasma of dog affected by leishmaniasis, the oxidative stress changes that are observed in this disease, are possibly mainly due to cellular blood components i.e., neutrophils responsible for the elimination of the parasite. Further studies are required to assess the clinical values of the collected data.
Collapse
|