1
|
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
|
2
|
Ahmad D, Linares I, Pietropaoli A, Waugh RE, McGrath JL. Sided Stimulation of Endothelial Cells Modulates Neutrophil Trafficking in an In Vitro Sepsis Model. Adv Healthc Mater 2024; 13:e2304338. [PMID: 38547536 PMCID: PMC11338706 DOI: 10.1002/adhm.202304338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Indexed: 04/09/2024]
Abstract
While the role of dysregulated polymorphonuclear leukocyte (PMN) transmigration in septic mediated tissue damage is well documented, strategies to mitigate aberrant transmigration across endothelium have yet to yield viable therapeutics. Recently, microphysiological systems (MPS) have emerged as novel in vitro mimetics that facilitate the development of human models of disease. With this advancement, aspects of endothelial physiology that are difficult to assess with other models can be directly probed. In this study, the role of endothelial cell (EC) apicobasal polarity on leukocyte trafficking response is evaluated with the µSiM-MVM (microphysiological system enabled by a silicon membrane - microvascular mimetic). Here, ECs are stimulated either apically or basally with a cytokine cocktail to model a septic-like challenge before introducing healthy donor PMNs into the device. Basally oriented stimulation generated a stronger PMN transmigratory response versus apical stimulation. Importantly, healthy PMNs are unable to migrate towards a bacterial peptide chemoattractant when ECs are apically stimulated, which mimics the attenuated PMN chemotaxis seen in sepsis. Escalating the apical inflammatory stimulus by a factor of five is necessary to elicit high PMN transmigration levels across endothelium. These results demonstrate that EC apicobasal polarity modulates PMN transmigratory behavior and provides insight into the mechanisms underlying sepsis.
Collapse
Affiliation(s)
- Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Isabelle Linares
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Anthony Pietropaoli
- Department of Medicine, Pulmonary Diseases and Critical Care at the University of Rochester, Rochester, NY, 14627, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| |
Collapse
|
3
|
Fowler EA, Farias Amorim C, Mostacada K, Yan A, Amorim Sacramento L, Stanco RA, Hales ED, Varkey A, Zong W, Wu GD, de Oliveira CI, Collins PL, Novais FO. Neutrophil-mediated hypoxia drives pathogenic CD8+ T cell responses in cutaneous leishmaniasis. J Clin Invest 2024; 134:e177992. [PMID: 38833303 PMCID: PMC11245163 DOI: 10.1172/jci177992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/17/2024] [Indexed: 06/06/2024] Open
Abstract
Cutaneous leishmaniasis caused by Leishmania parasites exhibits a wide range of clinical manifestations. Although parasites influence disease severity, cytolytic CD8+ T cell responses mediate disease. Although these responses originate in the lymph node, we found that expression of the cytolytic effector molecule granzyme B was restricted to lesional CD8+ T cells in Leishmania-infected mice, suggesting that local cues within inflamed skin induced cytolytic function. Expression of Blimp-1 (Prdm1), a transcription factor necessary for cytolytic CD8+ T cell differentiation, was driven by hypoxia within the inflamed skin. Hypoxia was further enhanced by the recruitment of neutrophils that consumed oxygen to produce ROS and ultimately increased the hypoxic state and granzyme B expression in CD8+ T cells. Importantly, lesions from patients with cutaneous leishmaniasis exhibited hypoxia transcription signatures that correlated with the presence of neutrophils. Thus, targeting hypoxia-driven signals that support local differentiation of cytolytic CD8+ T cells may improve the prognosis for patients with cutaneous leishmaniasis, as well as for other inflammatory skin diseases in which cytolytic CD8+ T cells contribute to pathogenesis.
Collapse
Affiliation(s)
- Erin A. Fowler
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Klauss Mostacada
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Allison Yan
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Rae A. Stanco
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Emily D.S. Hales
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Aditi Varkey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Wenjing Zong
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Camila I. de Oliveira
- Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Brazil
| | - Patrick L. Collins
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Fernanda O. Novais
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
4
|
Alonaizan R. Molecular regulation of NLRP3 inflammasome activation during parasitic infection. Biosci Rep 2024; 44:BSR20231918. [PMID: 38623843 PMCID: PMC11096646 DOI: 10.1042/bsr20231918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024] Open
Abstract
Parasitic diseases are a serious global health concern, causing many common and severe infections, including Chagas disease, leishmaniasis, and schistosomiasis. The NLRP3 inflammasome belongs to the NLR (nucleotide-binding domain leucine-rich-repeat-containing proteins) family, which are cytosolic proteins playing key roles in the detection of pathogens. NLRP3 inflammasomes are activated in immune responses to Plasmodium, Leishmania, Toxoplasma gondii, Entamoeba histolytica, Trypanosoma cruzi, and other parasites. The role of NLRP3 is not fully understood, but it is a crucial component of the innate immune response to parasitic infections and its functions as a sensor triggering the inflammatory response to the invasive parasites. However, while this response can limit the parasites' growth, it can also result in potentially catastrophic host pathology. This makes it essential to understand how NLRP3 interacts with parasites to initiate the inflammatory response. Plasmodium hemozoin, Leishmania glycoconjugate lipophosphoglycan (LPG) and E. histolytica Gal/GalNAc lectin can stimulate NLRP3 activation, while the dense granule protein 9 (GRA9) of T. gondii has been shown to suppress it. Several other parasitic products also have diverse effects on NLRP3 activation. Understanding the mechanism of NLRP3 interaction with these products will help to develop advanced therapeutic approaches to treat parasitic diseases. This review summarizes current knowledge of the NLRP3 inflammasome's action on the immune response to parasitic infections and aims to determine the mechanisms through which parasitic molecules either activate or inhibit its action.
Collapse
Affiliation(s)
- Rasha Alonaizan
- Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
5
|
La Rosa F, Varotto-Boccazzi I, Saresella M, Marventano I, Cattaneo GM, Hernis A, Piancone F, Otranto D, Epis S, Bandi C, Clerici M. The non-pathogenic protozoon Leishmania tarentolae interferes with the activation of NLRP3 inflammasome in human cells: new perspectives in the control of inflammation. Front Immunol 2024; 15:1298275. [PMID: 38707903 PMCID: PMC11066211 DOI: 10.3389/fimmu.2024.1298275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 04/03/2024] [Indexed: 05/07/2024] Open
Abstract
Background Innate immune responses against infectious agents can act as triggers of inflammatory diseases. On the other hand, various pathogens have developed mechanisms for the evasion of the immune response, based on an inhibition of innate immunity and inflammatory responses. Inflammatory diseases could thus be controlled through the administration of pathogens or pathogen-derived molecules, capable of interfering with the mechanisms at the basis of inflammation. In this framework, the NLRP3 inflammasome is an important component in innate antimicrobial responses and a major player in the inflammatory disease. Parasites of the genus Leishmania are master manipulators of innate immune mechanisms, and different species have been shown to inhibit inflammasome formation. However, the exploitation of pathogenic Leishmania species as blockers of NLRP3-based inflammatory diseases poses safety concerns. Methods To circumvent safety issues associated with pathogenic parasites, we focused on Leishmania tarentolae, a species of Leishmania that is not infectious to humans. Because NLRP3 typically develops in macrophages, in response to the detection and engulfment microorganisms, we performed our experiments on a monocyte-macrophage cell line (THP-1), either wild type or knockout for ASC, a key component of NLRP3 formation, with determination of cytokines and other markers of inflammation. Results L. tarentolae was shown to possess the capability of dampening the formation of NLRP3 inflammasome and the consequent expression of pro-inflammatory molecules, with minor differences compared to effects of pathogenic Leishmania species. Conclusion The non-pathogenic L. tarentolae appears a promising pro-biotic microbe with anti-inflammatory properties or a source of immune modulating cellular fractions or molecules, capable of interfering with the formation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences, University of Milan, Milan, Italy
- Pediatric Clinical Research Center 'Romeo ed Enrica Invernizzi', University of Milan, Milan, Italy
| | | | | | | | - Ambra Hernis
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | | | - Domenico Otranto
- Department of Veterinary Medicine, University of Bari, Valenzano, Italy
- Faculty of Veterinary Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Sara Epis
- Department of Biosciences, University of Milan, Milan, Italy
- Pediatric Clinical Research Center 'Romeo ed Enrica Invernizzi', University of Milan, Milan, Italy
| | - Claudio Bandi
- Department of Biosciences, University of Milan, Milan, Italy
- Pediatric Clinical Research Center 'Romeo ed Enrica Invernizzi', University of Milan, Milan, Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Jaimes J, Patiño LH, Herrera G, Cruz C, Pérez J, Correa-Cárdenas CA, Muñoz M, Ramírez JD. Prokaryotic and eukaryotic skin microbiota modifications triggered by Leishmania infection in localized Cutaneous Leishmaniasis. PLoS Negl Trop Dis 2024; 18:e0012029. [PMID: 38478569 PMCID: PMC10962849 DOI: 10.1371/journal.pntd.0012029] [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: 11/09/2023] [Revised: 03/25/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Cutaneous Leishmaniasis (CL) is a tropical disease characterized by cutaneous ulcers, sometimes with satellite lesions and nodular lymphangitis. Leishmania parasites, transmitted by sandfly vectors, cause this widespread public health challenge affecting millions worldwide. CL's complexity stems from diverse Leishmania species and intricate host interactions. Therefore, this study aims to shed light on the spatial-temporal distribution of Leishmania species and exploring the influence of skin microbiota on disease progression. We analyzed 40 samples from CL patients at three military bases across Colombia. Using Oxford Nanopore's Heat Shock Protein 70 sequencing, we identified Leishmania species and profiled microbiota in CL lesions and corresponding healthy limbs. Illumina sequencing of 16S-rRNA and 18S-rRNA genes helped analyze prokaryotic and eukaryotic communities. Our research uncovered a spatial-temporal overlap between regions of high CL incidence and our sampling locations, indicating the coexistence of various Leishmania species. L. naiffi emerged as a noteworthy discovery. In addition, our study delved into the changes in skin microbiota associated with CL lesions sampled by scraping compared with healthy skin sampled by brushing of upper and lower limbs. We observed alterations in microbial diversity, both in prokaryotic and eukaryotic communities, within the lesioned areas, signifying the potential role of microbiota in CL pathogenesis. The significant increase in specific bacterial families, such as Staphylococcaceae and Streptococcaceae, within CL lesions indicates their contribution to local inflammation. In essence, our study contributes to the ongoing research into CL, highlighting the need for a multifaceted approach to decipher the intricate interactions between Leishmaniasis and the skin microbiota.
Collapse
Affiliation(s)
- Jesús Jaimes
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Luz Helena Patiño
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Giovanny Herrera
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Claudia Cruz
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Julie Pérez
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Camilo A. Correa-Cárdenas
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
7
|
Ivănescu L, Andronic BL, Grigore-Hristodorescu S, Martinescu GV, Mîndru R, Miron L. The immune response in canine and human leishmaniasis and how this influences the diagnosis- a review and assessment of recent research. Front Cell Infect Microbiol 2023; 13:1326521. [PMID: 38149009 PMCID: PMC10749942 DOI: 10.3389/fcimb.2023.1326521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Leishmaniasis is a widespread but still underdiagnosed parasitic disease that affects both humans and animals. There are at least 20 pathogenic species of Leishmania, most of them being zoonotic. The diagnosis of leishmaniasis remains a major challenge, with an important role being played by the species of parasites involved, the genetic background, the immunocompetence of the host. This paper brings to the fore the sensitivity of the balance in canine and human leishmaniasis and addresses the importance of the host's immune response in establishing a correct diagnosis, especially in certain cases of asymptomatic leishmaniasis, or in the situation the host is immunosuppressed or acquired leishmaniasis through vertical transmission. The methods considered as a reference in the diagnosis of leishmaniasis no longer present certainty, the diagnosis being influenced mostly by the immune response of the host, which differs according to the presence of other associated diseases or even according to the breed in dogs. Consequently, the diagnosis and surveillance of leishmaniasis cases remains an open topic, requiring new diagnostic methods adapted to the immunological state of the host.
Collapse
Affiliation(s)
- Larisa Ivănescu
- Clinics Department, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Iaşi, Romania
| | - Bianca Lavinia Andronic
- Clinics Department, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Iaşi, Romania
| | | | | | - Raluca Mîndru
- Clinics Department, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Iaşi, Romania
| | - Liviu Miron
- Clinics Department, Faculty of Veterinary Medicine, Iasi University of Life Sciences, Iaşi, Romania
| |
Collapse
|
8
|
Amaral MP, Cardoso FD, de Farias IS, de Souza RQ, Matteucci KC, Torrecilhas AC, Bortoluci KR. NAIP/NLRC4 inflammasome participates in macrophage responses to Trypanosoma cruzi by a mechanism that relies on cathepsin-dependent caspase-1 cleavage. Front Immunol 2023; 14:1282856. [PMID: 38124741 PMCID: PMC10731265 DOI: 10.3389/fimmu.2023.1282856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Inflammasomes are large protein complexes that, once activated, initiate inflammatory responses by activating the caspase-1 protease. They play pivotal roles in host defense against pathogens. The well-established role of NAIP/NLRC4 inflammasome in bacterial infections involves NAIP proteins functioning as sensors for their ligands. However, recent reports have indicated the involvement of NLRC4 in non-bacterial infections and sterile inflammation, even though the role of NAIP proteins and the exact molecular mechanisms underlying inflammasome activation in these contexts remain to be elucidated. In this study, we investigated the activation of the NAIP/NLRC4 inflammasome in response to Trypanosoma cruzi, the protozoan parasite responsible for causing Chagas disease. This parasite has been previously demonstrated to activate NLRP3 inflammasomes. Here we found that NAIP and NLRC4 proteins are also required for IL-1β and Nitric Oxide (NO) release in response to T. cruzi infection, with their absence rendering macrophages permissive to parasite replication. Moreover, Nlrc4 -/- and Nlrp3 -/- macrophages presented similar impaired responses to T. cruzi, underscoring the non-redundant roles played by these inflammasomes during infection. Notably, it was the live trypomastigotes rather than soluble antigens or extracellular vesicles (EVs) secreted by them, that activated inflammasomes in a cathepsins-dependent manner. The inhibition of cathepsins effectively abrogated caspase-1 cleavage, IL-1β and NO release, mirroring the phenotype observed in Nlrc4 -/-/Nlrp3 -/- double knockout macrophages. Collectively, our findings shed light on the pivotal role of the NAIP/NLRC4 inflammasome in macrophage responses to T. cruzi infection, providing new insights into its broader functions that extend beyond bacterial infections.
Collapse
Affiliation(s)
- Marcelo Pires Amaral
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Felipe Daniel Cardoso
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Ingrid Sancho de Farias
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Rafael Queiroz de Souza
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Kely Catarine Matteucci
- Plataforma de Medicina Translacional, Fundação Oswaldo Cruz (FIOCRUZ), Faculdade de Medicina de Ribeirão Preto (FMRP), Ribeirão Preto, SP, Brazil
| | - Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Karina Ramalho Bortoluci
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
9
|
Singh TP, Farias Amorim C, Lovins VM, Bradley CW, Carvalho LP, Carvalho EM, Grice EA, Scott P. Regulatory T cells control Staphylococcus aureus and disease severity of cutaneous leishmaniasis. J Exp Med 2023; 220:e20230558. [PMID: 37812390 PMCID: PMC10561556 DOI: 10.1084/jem.20230558] [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: 04/03/2023] [Revised: 08/02/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
Cutaneous leishmaniasis causes alterations in the skin microbiota, leading to pathologic immune responses and delayed healing. However, it is not known how these microbiota-driven immune responses are regulated. Here, we report that depletion of Foxp3+ regulatory T cells (Tregs) in Staphylococcus aureus-colonized mice resulted in less IL-17 and an IFN-γ-dependent skin inflammation with impaired S. aureus immunity. Similarly, reducing Tregs in S. aureus-colonized and Leishmania braziliensis-infected mice increased IFN-γ, S. aureus, and disease severity. Importantly, analysis of lesions from L. braziliensis patients revealed that low FOXP3 gene expression is associated with high IFNG expression, S. aureus burden, and delayed lesion resolution compared to patients with high FOXP3 expression. Thus, we found a critical role for Tregs in regulating the balance between IL-17 and IFN-γ in the skin, which influences both bacterial burden and disease. These results have clinical ramifications for cutaneous leishmaniasis and other skin diseases associated with a dysregulated microbiome when Tregs are limited or dysfunctional.
Collapse
Affiliation(s)
- Tej Pratap Singh
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camila Farias Amorim
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria M. Lovins
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles W. Bradley
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lucas P. Carvalho
- Servico de Imunologia, Complexo Hospitalar Universitario Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratorio de Pesquisas Clinicas do Instituto de Pesquisas Goncalo Moniz, Fiocruz, Salvador, Brazil
| | - Edgar M. Carvalho
- Servico de Imunologia, Complexo Hospitalar Universitario Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratorio de Pesquisas Clinicas do Instituto de Pesquisas Goncalo Moniz, Fiocruz, Salvador, Brazil
| | - Elizabeth A. Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Fowler EA, Amorim CF, Mostacada K, Yan A, Sacramento LA, Stanco RA, Hales EDS, Varkey A, Zong W, Wu GD, de Oliveira CI, Collins PL, Novais FO. Pathogenic CD8 T cell responses are driven by neutrophil-mediated hypoxia in cutaneous leishmaniasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562926. [PMID: 37904953 PMCID: PMC10614852 DOI: 10.1101/2023.10.18.562926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Cutaneous leishmaniasis caused by Leishmania parasites exhibits a wide range of clinical manifestations. Although parasites influence disease severity, cytolytic CD8 T cell responses mediate disease. While these responses originate in the lymph node, we find that expression of the cytolytic effector molecule granzyme B is restricted to lesional CD8 T cells in Leishmania - infected mice, suggesting that local cues within inflamed skin induce cytolytic function. Expression of Blimp-1 ( Prdm1 ), a transcription factor necessary for cytolytic CD8 T cell differentiation, is driven by hypoxia within the inflamed skin. Hypoxia is further enhanced by the recruitment of neutrophils that consume oxygen to produce reactive oxygen species, ultimately increasing granzyme B expression in CD8 T cells. Importantly, lesions from cutaneous leishmaniasis patients exhibit hypoxia transcription signatures that correlate with the presence of neutrophils. Thus, targeting hypoxia-driven signals that support local differentiation of cytolytic CD8 T cells may improve the prognosis for patients with cutaneous leishmaniasis, as well as other inflammatory skin diseases where cytolytic CD8 T cells contribute to pathogenesis.
Collapse
|
11
|
Sengupta R, Roy M, Dey NS, Kaye PM, Chatterjee M. Immune dysregulation and inflammation causing hypopigmentation in post kala-azar dermal leishmaniasis: partners in crime? Trends Parasitol 2023; 39:822-836. [PMID: 37586987 DOI: 10.1016/j.pt.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Post kala-azar dermal leishmaniasis (PKDL), a heterogeneous dermal sequela of visceral leishmaniasis (VL), is challenging in terms of its etiopathogenesis. Hypopigmentation is a consistent clinical feature in PKDL, but mechanisms contributing to the loss of melanocytes remains poorly defined. Like other hypopigmentary dermatoses - for example, vitiligo, psoriasis, and leprosy - the destruction of melanocytes is likely a multifactorial phenomenon, key players being immune dysregulation and inflammation. This review focuses on immunological mechanisms responsible for the 'murder' of melanocytes, prime suspects at the lesional sites being CD8+ T cells and keratinocytes and their criminal tools being proinflammatory cytokines, for example, IFN-γ, IL-6, and TNF-α. Collectively, these may cause decreased secretion of melanocyte growth factors, loss/attenuation of cell adhesion molecules and inflammasome activation, culminating in melanocyte death.
Collapse
Affiliation(s)
- Ritika Sengupta
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B AJC Bose Road, Kolkata 700020, India
| | - Madhurima Roy
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B AJC Bose Road, Kolkata 700020, India
| | - Nidhi S Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York, YO10 5DD, UK
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York, YO10 5DD, UK
| | - Mitali Chatterjee
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research, 244B AJC Bose Road, Kolkata 700020, India.
| |
Collapse
|
12
|
Mor B, Görmez A, Demirci B. Immunopathological investigation of a gerbil model of cutaneous leishmaniasis. Acta Trop 2023; 246:106991. [PMID: 37479161 DOI: 10.1016/j.actatropica.2023.106991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/07/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Leishmaniasis, caused by Leishmania species (intracellular protozoans), is a chronic, systemic disease that causes skin (cutaneous) and internal organ infections (visceral). Its prevalence has increased in recent years. Leishmania species are considered important pathogens that affect public health. After infecting an individual, the pathogen disrupts the immune system, but, there are not enough studies on which immune mechanisms are affected. The aim of this study was to establish a Leishmania major infection model (the causative agent of cutaneous leishmaniasis) in gerbils (Meriones unguiculatus) and to investigate the immune response in this model by examining the expression of important inflammatory genes (IL-1β, IL-2, IL-6, IFN-ɣ and TNF-α). The presence of parasites was confirmed by microscopic examination of samples taken from the lesions and culture studies. The expression of inflammatory cytokine genes was significantly increased in infected gerbils. The changes indicated that both the Th1 and Th2 pathways are activated in cutaneous leishmaniasis infection. Hence, different immunopathological mechanisms should be evaluated in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Baycan Mor
- Department of Molecular Biology and Genetics, Faculty of Science, Kafkas University, 36100, Kars, Türkiye.
| | - Arzu Görmez
- Department of Biology, Faculty of Science, Dokuz Eylul University, 35390, Izmir, Turkey
| | - Berna Demirci
- Department of Molecular Biology and Genetics, Faculty of Science, Kafkas University, 36100, Kars, Türkiye
| |
Collapse
|
13
|
DeMichele E, Sosnowski O, Buret AG, Allain T. Regulatory Functions of Hypoxia in Host-Parasite Interactions: A Focus on Enteric, Tissue, and Blood Protozoa. Microorganisms 2023; 11:1598. [PMID: 37375100 PMCID: PMC10303274 DOI: 10.3390/microorganisms11061598] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Body tissues are subjected to various oxygenic gradients and fluctuations and hence can become transiently hypoxic. Hypoxia-inducible factor (HIF) is the master transcriptional regulator of the cellular hypoxic response and is capable of modulating cellular metabolism, immune responses, epithelial barrier integrity, and local microbiota. Recent reports have characterized the hypoxic response to various infections. However, little is known about the role of HIF activation in the context of protozoan parasitic infections. Growing evidence suggests that tissue and blood protozoa can activate HIF and subsequent HIF target genes in the host, helping or hindering their pathogenicity. In the gut, enteric protozoa are adapted to steep longitudinal and radial oxygen gradients to complete their life cycle, yet the role of HIF during these protozoan infections remains unclear. This review focuses on the hypoxic response to protozoa and its role in the pathophysiology of parasitic infections. We also discuss how hypoxia modulates host immune responses in the context of protozoan infections.
Collapse
Affiliation(s)
- Emily DeMichele
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
14
|
Sequera HDG, de Souza JL, Junior JDES, da Silva LS, Pinheiro SK, Kerr HKA, de Souza MLG, Guerra MVDF, de Mesquita TGR, Ramasawmy R. Variants of CARD8 in Leishmania guyanensis-cutaneous leishmaniasis and influence of the variants genotypes on circulating plasma cytokines IL-1β, TNFα and IL-8. PLoS Negl Trop Dis 2023; 17:e0011416. [PMID: 37276232 PMCID: PMC10270566 DOI: 10.1371/journal.pntd.0011416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 06/15/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023] Open
Abstract
Nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein family (NLR) are intracellular pathogen recognition receptors mediating innate immunity, releasing proinflammatory cytokines IL-1β and IL-18, and promoting pyroptotic cell death, upon sensing pathogenic or endogenous danger signals. In animal models, NLRP3 inflammasome has a dual role, pathogenic or protective in Leishmania-infection, depending on the Leishmania species and mice strain. Caspase recruitment containing domain 8 (CARD8) is a negative regulator of NLRP3 inflammasome and also an inhibitor of transcription factor NFĸB, a major transcription factor of proinflammatory cytokines. We investigated whether single nucleotide variants in CARD8 may partially explain why only a proportion of individuals coming from the same area of endemicity of leishmaniasis develop cutaneous leishmaniasis caused by Leishmania guyanensis. We genotyped four single nucleotide variants of the CARD8 gene by direct nucleotide sequencing in 1741 individuals from an endemic area of leishmaniasis, constituting 850 patients with CL and 891 healthy controls. The frequencies of the genotypes of the variants rs2288877 T>C, rs73944113 C>T, and rs2043211 A>T are similar among the patients with CL and HC, while the variant rs2288876 A>G) reveals an excess of the genotype AA among the patients with CL (44%) compared to 37% in the HC group. Allele A of the variant rs2288876 A>G) is associated with susceptibility to CL (OR = 1.2 [95%CI 1.03-1.4]; P = 0.01). Haplotype analysis showed that individuals harboring the haplotype CCAA have 280% odds of developing CL caused by L. guyanensis (OR = 3.8 [95% CI 2.0-7.7]; p = 0.00004). The variants rs2288877 T>C and rs2288876 A>G correlate with the plasma level of IL-8. Spearman correlation showed a significant positive correlation between the rs2288876 A>G allele A and the level of IL-8 (ρ = 0.22; p = 0.0002). CARD8 may partially contribute to the development of CL caused by L. guyanensis.
Collapse
Affiliation(s)
| | - Josué Lacerda de Souza
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus, Brazil
| | - José do Espírito Santo Junior
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas, Manaus, Brazil
| | - Lener Santos da Silva
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
| | - Suzana Kanawati Pinheiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
| | | | | | - Marcus Vinitius de Farias Guerra
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
| | - Tirza Gabrielle Ramos de Mesquita
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
| | - Rajendranath Ramasawmy
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Faculdade de Medicina Nilton Lins, Universidade Nilton Lins, Manaus, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazonia Legal (Rede Bionorte), Universidade do Estado do Amazonas, Manaus, Brazil
- Fundação de Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Brazil
- Genomic Health Surveillance Network: Optimization of Assistance and Research in the State of Amazonas (REGESAM), Manaus, Brazil
| |
Collapse
|
15
|
Goto Y, Mizobuchi H. Pathological roles of macrophages in Leishmania infections. Parasitol Int 2023; 94:102738. [PMID: 36738983 DOI: 10.1016/j.parint.2023.102738] [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: 10/27/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Macrophages are the major host cells for Leishmania parasites, and determine the fate of infection by either limiting or allowing growth of the parasites, resulting in development or control of leishmaniasis, respectively. They also play important roles in causing pathological outcomes during Leishmania infection. The pathophysiology is complex and include a wide variety of molecular and cellular responses including enhancement of inflammatory responses by releasing cytokines, causing damages to surrounding cells by reactive oxygen species, or disordered phagocytosis of other cells. It is of note that disease severity in leishmaniasis sometimes does not correlate with parasite burdens, indicating that pathological roles of macrophages are not necessarily linked to their parasite-killing activities that are often defined by M1/M2 status. Here, we review the roles of macrophages in leishmaniasis with a focus on their pathological mechanisms in disease development.
Collapse
Affiliation(s)
- Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | - Haruka Mizobuchi
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
16
|
Traughber CA, Iacano AJ, Neupane K, Khan MR, Opoku E, Nunn T, Prince A, Sangwan N, Hazen SL, Smith JD, Gulshan K. Impavido attenuates inflammation, reduces atherosclerosis, and alters gut microbiota in hyperlipidemic mice. iScience 2023; 26:106453. [PMID: 37020959 PMCID: PMC10067757 DOI: 10.1016/j.isci.2023.106453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/14/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Impavido (Miltefosine) is an FDA-approved drug for treating leishmaniasis and primary amebic meningoencephalitis. We have shown previously that Miltefosine increased cholesterol release and dampened Nlrp3 inflammasome assembly in macrophages. Here, we show that Miltefosine reduced LPS-induced choline uptake by macrophages, and attenuated Nlrp3 inflammasome assembly in mice. Miltefosine-fed mice showed reduced plasma IL-1β in a polymicrobial cecal slurry model of systemic inflammation. Miltefosine-fed mice showed increased reverse cholesterol transport to the plasma, liver, and feces. Hyperlipidemic apoE-/- mice fed with WTD + Miltefosine showed significantly reduced weight gain and markedly reduced atherosclerotic lesions versus mice fed with WTD. The 16S rDNA sequencing and analysis of gut microbiota showed marked alterations in the microbiota profile of Miltefosine-fed hyperlipidemic apoE-/- versus control, with the most notable changes in Romboutsia and Bacteriodes species. Taken together, these data indicate that Miltefosine causes pleiotropic effects on lipid metabolism, inflammasome activity, atherosclerosis, and the gut microbiota.
Collapse
|
17
|
Zhu F, Yu D, Qin X, Qian Y, Ma J, Li W, Liu Q, Wang C, Zhang Y, Li Y, Jiang D, Wang S, Xia P. The neuropeptide CGRP enters the macrophage cytosol to suppress the NLRP3 inflammasome during pulmonary infection. Cell Mol Immunol 2023; 20:264-276. [PMID: 36600053 PMCID: PMC9970963 DOI: 10.1038/s41423-022-00968-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 12/11/2022] [Indexed: 01/06/2023] Open
Abstract
The NLRP3 inflammasome plays an essential role in resistance to bacterial infection. The nervous system secretes multiple neuropeptides affecting the nervous system as well as immune cells. The precise impact of the neuropeptide CGRP on NLRP3 inflammasome activation is still unclear. Here, we show that CGRP negatively regulates the antibacterial process of host cells. CGRP prevents NLRP3 inflammasome activation and reduces mature IL-1β secretion. Following NLRP3 inflammasome stimulation that triggers endosome leakage, CGRP internalized to endosomal compartments is released into the cell cytosol. Cytosolic CGRP binds directly to NLRP3 and dismantles the NLRP3-NEK7 complex, which is crucial for NLRP3 inflammasome activation. CGRP administration exacerbates bacterial infection, while the treatment with a CGRP antagonist has the opposite effect. Our study uncovers a unique role of CGRP in inhibiting inflammasome activation during infections, which might shed new light on antibacterial therapies in the future.
Collapse
Affiliation(s)
- Fangrui Zhu
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Dou Yu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Xiwen Qin
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Yan Qian
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Juan Ma
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Weitao Li
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Qiannv Liu
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Chunlei Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Yan Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China
| | - Yi Li
- Department of Anesthesiology, Peking University Third Hospital, 100191, Beijing, China
| | - Dong Jiang
- Department of Sports Medicine, Peking University Third Hospital, 100191, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine of Peking University, 100191, Beijing, China
| | - Shuo Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences, Peking University, 100191, Beijing, China.
- NHC Key Laboratory of Medical Immunology, Peking University, 100191, Beijing, China.
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, 100191, Beijing, China.
| |
Collapse
|
18
|
Rahmanipour M, Mohebali M, Koosha M, Kazemirad E, Yasami-Khiabani S, Mirjalali H, Hajjaran H. Effect of Leishmania RNA virus 2 on virulence factors and cytokines gene expression in a human macrophage infected with Leishmania major: A preliminary study. Exp Parasitol 2023; 246:108459. [PMID: 36596336 DOI: 10.1016/j.exppara.2022.108459] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023]
Abstract
Cutaneous leishmaniasis (CL) is one of the most important infectious parasitic diseases in the world caused by the Leishmania parasite. In recent decades, the presence of a virus from the Totiviridae family has been proven in some Leishmania species. Although the existence of LRV2 in the Old world Leishmania species has been confirmed, almost no studies have been done to determine the potential impact of LRV2 on the immunopathogenicity of the Leishmania parasite. In this preliminary study, we measured the expression of target genes, including Glycoprotein 63 (gp63), Heat Shock Protein 70 (hsp70), Cysteine Protease b (cpb), Interleukin 1 beta (IL-1β), IL8 and IL-12 in LRV2 positive Leishmania major strain (LRV2+L. major) and LRV2 negative L. major strain (LRV2-L. major). We exposed THP-1, a human leukemia monocytic cell line, to promastigotes of both strains. After the initial infection, RNA was extracted at different time points, and the relative gene expression was determined using a real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Findings showed that the presence of LRV2 in L. major was able to increase the expression of gp63, hsp70, and cpb genes; also, we observed lower levels of expression in cytokine genes of IL-1β, IL-8, IL-12 in the presence of LRV2+, which are critical factors in the host's immune response against leishmaniasis. These changes could suggest that the presence of LRV2 in L. major parasite may change the outcome of the disease and increase the probability of Leishmania survival; nevertheless, further studies are needed to confirm our results.
Collapse
Affiliation(s)
- Milad Rahmanipour
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Center for Research of Endemic Parasites of Iran (CREPI), Tehran University of Medical Sciences, Tehran, Iran.
| | - Mona Koosha
- Department of Medical Entomology & Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Kazemirad
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homa Hajjaran
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
19
|
de Sá KSG, Amaral LA, Rodrigues TS, Ishimoto AY, de Andrade WAC, de Almeida L, Freitas-Castro F, Batah SS, Oliveira SC, Pastorello MT, Fabro AT, Zamboni DS. Gasdermin-D activation promotes NLRP3 activation and host resistance to Leishmania infection. Nat Commun 2023; 14:1049. [PMID: 36828815 PMCID: PMC9958042 DOI: 10.1038/s41467-023-36626-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
Intracellular parasites from the Leishmania genus cause Leishmaniasis, a disease affecting millions of people worldwide. NLRP3 inflammasome is key for disease outcome, but the molecular mechanisms upstream of the inflammasome activation are still unclear. Here, we demonstrate that despite the absence of pyroptosis, Gasdermin-D (GSDMD) is active at the early stages of Leishmania infection in macrophages, allowing transient cell permeabilization, potassium efflux, and NLRP3 inflammasome activation. Further, GSDMD is processed into a non-canonical 25 kDa fragment. Gsdmd-/- macrophages and mice exhibit less NLRP3 inflammasome activation and are highly susceptible to infection by several Leishmania species, confirming the role of GSDMD for inflammasome-mediated host resistance. Active NLRP3 inflammasome and GSDMD are present in skin biopsies of patients, demonstrating activation of this pathway in human leishmaniasis. Altogether, our findings reveal that Leishmania subverts the normal functions of GSDMD, an important molecule to promote inflammasome activation and immunity in Leishmaniasis.
Collapse
Affiliation(s)
- Keyla S G de Sá
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luana A Amaral
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Tamara S Rodrigues
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Adriene Y Ishimoto
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Warrison A C de Andrade
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Leticia de Almeida
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Felipe Freitas-Castro
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sabrina S Batah
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Mônica T Pastorello
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Alexandre T Fabro
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
- Serviço de Patologia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
20
|
Jawale D, Khandibharad S, Singh S. Decoding systems immunological model of sphingolipids with IL-6/IL-17/IL-23 axes in L. major infection. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159261. [PMID: 36494028 DOI: 10.1016/j.bbalip.2022.159261] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
IL-6, IL-17, IL-23 and IL-1β are the crucial cytokines controlling inflammatory and immune response during L. major infection. During cutaneous leishmaniasis, an important T helper cell type CD4+ Th17 subset plays a deterministic role in lesion formation through channelling infected macrophages and production of IL-1β, IL-6, IL-23 and IFN-γ. Ceramide derived sphingosine precursors may assist in pro-inflammatory cytokine response. However, the role of these metabolites in inflammation with pleiotropic pro-inflammatory cytokines in L. major infection is unknown. The present study indicates IL-6/IL-17/IL-23 and SPHK1-S1P-S1PRs signaling axes with the overexpression of SATB1 aiding in disease progression. Targeting SATB1 might modulate the secretion of pro-inflammatory cytokines and abnormal immune functioning, thereby killing the intracellular parasite. Systems immunological methods assisted in a step towards identifying the key to the mystery of crucial components and serving as an approach for therapeutic intervention in L. major infection.
Collapse
Affiliation(s)
- Diksha Jawale
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Shweta Khandibharad
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India.
| |
Collapse
|
21
|
Infection and Immunity. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
22
|
Hong Z, Cheng J, Ye Y, Chen X, Zhang F. MicroRNA-451 Attenuates the Inflammatory Response of Activated Microglia by Downregulating Nucleotide Binding Oligomerization Domain-Like Receptor Protein 3. World Neurosurg 2022; 167:e1128-e1137. [PMID: 36087911 DOI: 10.1016/j.wneu.2022.08.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Spinal cord injury is the most common problem encountered during spinal surgery. After the initial trauma, the disruption of the blood-brain barrier and subsequent microglia activation result in extensive inflammatory responses. Inflammasomes are large protein complexes that are essential during inflammation. One of the most studied inflammasome components, nucleotide binding oligomerization domain-like receptor protein 3 (NLRP; nucleotide binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing 3), is widely expressed in the central nervous system. Previous research has shown that microRNA-451 (miR-451) might play a role in regulating inflammatory conditions. METHODS Using bioinformatics analysis, we found that NLRP3 is a direct target of miR-451. This in silico prediction was confirmed using dual-luciferase reporter gene assays. To further demonstrate that miR-451 influenced microglial NLRP3 production, we activated microglial cells with lipopolysaccharides. RESULTS Activating microglial cells with lipopolysaccharides resulted in the production of NLRP3 inflammasomes and the secretion of the proinflammatory cytokines interleukin-1β and interleukin-18. We were able to demonstrate that overexpression of miR-451 suppressed this NLRP3-induced proinflammatory cascade of events. CONCLUSIONS Our findings have highlighted the potential anti-inflammatory role of miR-451 in reducing the secondary neuronal damage after spinal cord injury.
Collapse
Affiliation(s)
- Zhou Hong
- Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaqi Cheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Yong Ye
- Medical School of Nantong University, Nantong, China
| | - Xiaoqing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
23
|
Leishmania major Strain-Dependent Macrophage Activation Contributes to Pathogenicity in the Absence of Lymphocytes. Microbiol Spectr 2022; 10:e0112622. [PMID: 36190414 PMCID: PMC9603372 DOI: 10.1128/spectrum.01126-22] [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] [Indexed: 12/30/2022] Open
Abstract
Infection of C57BL/6 wild-type mice with Leishmania major 5-ASKH or Friedlin strains results in relatively similar pathogenicity with self-healing lesions within weeks. Parasite clearance depends on nitric oxide production by activated macrophages in response to cytokines produced mainly by CD4+ Th1 cells. In contrast, C57BL/6 Rag2 knockout mice, which lack T and B lymphocytes, show distinct pathologies during infection with these strains. Despite of the similar parasite number, the 5-ASKH infection induced severe inflammation rather than the Friedlin. To determine the immunological factors behind this phenomenon, we infected C57BL/6 Rag2 knockout mice with these two strains and compared immune cell kinetics and macrophage activation status. Compared with the Friedlin strain, the 5-ASKH strain elicited increased pathology associated with the accumulation of CD11bhigh, Ly6Ghigh neutrophils by week four and increased the expression of macrophage activation markers. We then analyzed the differentially expressed transcripts in infected bone marrow-derived macrophages by RNA sequencing. It showed upregulation of multiple inflammatory transcripts, including Toll-like receptor 1/2 (TLR1/2), CD69, and CARD14, upon 5-ASKH infection. Our findings suggest that different L. major strains can trigger distinct macrophage activation, contributing to the disease outcome observed in the absence of lymphocytes but not in the presence of lymphocytes. IMPORTANCE Disease manifestations of cutaneous leishmaniasis (CL) range from self-healing cutaneous lesions to chronic forms of the disease, depending on the infecting Leishmania sp. and host immune protection. Previous works on mouse models of CL show the distinct pathogenicity of Leishmania major strains in the absence of lymphocytes. However, the mechanisms of this pathology remain uncovered. In the trial to understand the immunological process involved in lymphocyte-independent pathology, we have found a specific induction of macrophages by different L. major strains that affect their ability to mount innate responses leading to neutrophilic pathology when lymphocytes are ablated.
Collapse
|
24
|
Abstract
Cytokines play crucial roles in commencing and coordinating the organized recruitment and activation of immune cells during infection. These molecular regulators play an important part in deciding the fate of disease outcomes in leishmaniasis, a parasitic disease of tropical and subtropical countries. T helper 1 (Th1) cell-mediated inflammatory cytokines usually play a host-protective role, while T helper 2 (Th2) cell activation produces an anti-inflammatory milieu necessary for parasite survival. It is noteworthy that in such a multifaceted disease, the role played by any particular cytokine cannot be generalized as either beneficial or detrimental. For example, a "host-favorable" cytokine in one form of the disease has been found to be "pathogen friendly" in another form of leishmaniasis. On the other hand, the complex signaling network regulating the production of cytokines is further complicated by the nature of the host as well as the presence of other cytokines in the milieu. The present review focuses on the differential roles played by cytokines and the intricate signaling network responsible for the regulation of such cytokines during infection by different species of Leishmania. While many more studies are required in the future to better understand the role of these molecules in both animal models and patient samples, current studies indicate that these molecules are potential candidates to be targeted for therapy against this deadly disease.
Collapse
|
25
|
Gonçalves-de-Albuquerque SDC, da Silva LG, Sousa-Paula LCD, Sales KGDS, Boegel A, Dantas-Torres F. Exploring IL-17 gene promoter polymorphisms in canine leishmaniasis. Acta Trop 2022; 232:106452. [PMID: 35421384 DOI: 10.1016/j.actatropica.2022.106452] [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: 10/23/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022]
Abstract
Proinflammatory cytokine secretion determines the infection course in leishmaniasis. The immunopathology of canine leishmaniasis (CanL) caused by Leishmania infantum is characterized by low Leishmania-specific IFN-γ and IL-17 production. Mutations in the human IL-17 gene promoter alter cytokine expression and may increase the susceptibility of humans to some infectious diseases. In this study, we correlated canine IL-17 single nucleotide polymorphisms (SNPs) with anti-Leishmania IgG levels, parasite load and external clinical signs in dogs naturally exposed to L. infantum in Brazil. A higher frequency (Chi-square test: X2= 5.378, df= 1, P= 0.020) of major alleles was observed among dogs showing no external clinical signs attributable to Leishmania infection. A high proportion of A allele carriers (mutant) were observed among dogs with high antibody levels, although differences were not statistically significant (Chi-square test: X2= 4.410, df= 4, P= 0.353), as compared to dogs with low antibody levels. In general, the association of canine IL-17 SNPs with disease expression or disease exasperation did not reach enough statistical power to allow the use of these mutations as prognostic markers. This knowledge may pave the way for further investigations on the genetic aspects of CanL and its immunotherapy.
Collapse
Affiliation(s)
- Suênia da C Gonçalves-de-Albuquerque
- Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil; Central Laboratory of Public Health Dr Milton Bezerra de Sobral, Recife, Pernambuco, Brazil.
| | | | | | | | | | - Filipe Dantas-Torres
- Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil.
| |
Collapse
|
26
|
Muñoz-Durango N, Gómez A, García-Valencia N, Roldán M, Ochoa M, Bautista-Erazo DE, Ramírez-Pineda JR. A Mouse Model of Ulcerative Cutaneous Leishmaniasis by Leishmania (Viannia) panamensis to Investigate Infection, Pathogenesis, Immunity, and Therapeutics. Front Microbiol 2022; 13:907631. [PMID: 35770175 PMCID: PMC9234518 DOI: 10.3389/fmicb.2022.907631] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
A mouse model of cutaneous leishmaniasis (CL) by Leishmania (Viannia) panamensis (L(V)p) that reproduces the characteristics of the human disease remains elusive. Here we report the development of a CL model that uses a mouse-adapted L(V)p isolate to reproducibly induce a dermal disease with a remarkable similarity to human CL. BALB/c mice infected intradermally in the ear with 105 stationary UA-946 L(V)p promastigotes develop a progressive cutaneous disease that exhibits the typical ulcerated lesions with indurated borders observed in CL patients. Although most of parasites in the inoculum die within the first week of infection, the survivors vigorously multiply at the infection site during the following weeks, paralleling disease appearance and aggravation. Regional lymphadenopathy as well as lymphatic dissemination of parasites to draining lymph nodes (dLN) was evidenced early after infection. Viable parasites were also isolated from spleen at later timepoints indicating systemic parasitic dissemination, but, strikingly, no signs of systemic disease were observed. Increasing numbers of myeloid cells and T lymphocytes producing IFNγ and IL-4 were observed in the dLN as disease progressed. A mixed adaptive L(V)p-specific T cell-mediated response was induced, since ex vivo recall experiments using dLN cells and splenocytes revealed the production of type 1 (IFNγ, IL-2), type 2 (IL-4, IL-13), regulatory (IL-10), and inflammatory (GM-CSF, IL-3) cytokines. Humoral adaptive response was characterized by early production of IgG1- followed by IgG2a-type of L(V)p-specific antibodies. IFNγ/IL-4 and IgG2a/IgG1 ratios indicated that the initial non-protective Th2 response was redirected toward a protective Th1 response. In situ studies revealed a profuse recruitment of myeloid cells and of IFNγ- and IL-4-producing T lymphocytes to the site of infection, and the typical histopathological changes induced by dermotropic Leishmania species. Evidence that this model is suitable to investigate pharmacological and immunomodulatory interventions, as well as for antigen discovery and vaccine development, is also presented. Altogether, these results support the validity and utility of this novel mouse model to study the pathogenesis, immunity, and therapeutics of L(V)p infections.
Collapse
Affiliation(s)
- Natalia Muñoz-Durango
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Alexander Gómez
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Natalia García-Valencia
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Miguel Roldán
- Instituto de Patología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Marcela Ochoa
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - David E. Bautista-Erazo
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - José R. Ramírez-Pineda
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
27
|
DeSouza-Vieira T. The metamorphosis of neutrophil transcriptional program during Leishmania infection. Parasite Immunol 2022; 44:e12922. [PMID: 35437801 DOI: 10.1111/pim.12922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/30/2022] [Accepted: 04/14/2022] [Indexed: 11/27/2022]
Abstract
The role of neutrophils in the course of Leishmania infection remains controversial, displaying tremendous variability depending on the species of parasite, stage of infection, host genetic background, and methodological discrepancies among studies. Although neutrophils have long been categorized as short-lived cells with limited capacity to express proteins de novo, recent advances have revealed significant plasticity in neutrophil transcriptional programs and intrapopulation heterogeneity, which can be regulated by both intrinsic and extrinsic factors that together determine the profile of neutrophil effector response. In this review, we focus on the current understanding of neutrophil transcriptional plasticity, neutrotime, evidence of Leishmania-mediated alterations in neutrophil transcriptome leading to the rise of subpopulations, and finally, functional implications of those findings to the course of Leishmania infection.
Collapse
Affiliation(s)
- Thiago DeSouza-Vieira
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
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: 34] [Impact Index Per Article: 17.0] [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
|
29
|
Sanches RCO, Mambelli F, Oliveira SC. Neutrophils and schistosomiasis: a missing piece in pathology. Parasite Immunol 2022; 44:e12916. [PMID: 35332932 DOI: 10.1111/pim.12916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022]
Abstract
Schistosomiasis is a chronic human parasitic disease that causes serious health problems worldwide. The disease-associated liver pathology is one of the hallmarks of infections by S. mansoni and S. japonicum, and is accountable for the debilitating condition found in infected patients. In the past few years, investigative studies have highlighted the key role played by neutrophils and the influence of inflammasome signaling pathway in different pathological conditions. However, it is noteworthy that the study of inflammasome activation in neutrophils has been overlooked by reports concerning macrophages and monocytes. This interplay between neutrophils and inflammasomes is much more poorly investigated during schistosomiasis. Herein we reviewed the role of neutrophils during schistosomiasis and addressed the potential connection between these cells and inflammasome activation in this context.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio Mambelli
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, Salvador, Brazil
| |
Collapse
|
30
|
Persistent Cutaneous Leishmania major Infection Promotes Infection-Adapted Myelopoiesis. Microorganisms 2022; 10:microorganisms10030535. [PMID: 35336108 PMCID: PMC8954948 DOI: 10.3390/microorganisms10030535] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/11/2022] [Accepted: 02/26/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem/progenitor cells (HSPC) are responsible for the generation of most immune cells throughout the lifespan of the organism. Inflammation can activate bone marrow HSPCs, leading to enhanced myelopoiesis to replace cells, such as neutrophils, which are attracted to inflamed tissues. We have previously shown that HSPC activation promotes parasite persistence and expansion in experimental visceral leishmaniasis through the increased production of permissive monocytes. However, it is not clear if the presence of the parasite in the bone marrow was required for infection-adapted myelopoiesis. We therefore hypothesized that persistent forms of Leishmania major (cutaneous leishmaniasis) could also activate HSPCs and myeloid precursors in the C57Bl/6 mouse model of intradermal infection in the ear. The accrued influx of myeloid cells to the lesion site corresponded to an increase in myeloid-biased HSPCs in the bone marrow and spleen in mice infected with a persistent strain of L. major, together with an increase in monocytes and monocyte-derived myeloid cells in the spleen. Analysis of the bone marrow cytokine and chemokine environment revealed an attenuated type I and type II interferon response in the mice infected with the persistent strain compared to the self-healing strain, while both strains induced a rapid upregulation of myelopoietic cytokines, such as IL-1β and GM-CSF. These results demonstrate that an active infection in the bone marrow is not necessary for the induction of infection-adapted myelopoiesis, and underline the importance of considering alterations to the bone marrow output when analyzing in vivo host-pathogen interactions.
Collapse
|
31
|
Trifluoro-icaritin alleviates chronic inflammatory pain through α7nAChR-mediated suppression of HMGB1/NF-κB signaling in the spinal cord of rats. Brain Res Bull 2022; 183:13-26. [PMID: 35202753 DOI: 10.1016/j.brainresbull.2022.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 01/13/2022] [Accepted: 02/20/2022] [Indexed: 01/02/2023]
Abstract
Inflammatory pain is a chronic, persistent and serious disease that greatly impacts public health, which is often accompanied by allodynia, hyperalgesia, and spontaneous pain. It is evident that α7 nicotinic acetylcholine receptor (α7nAChR) plays a key role in cholinergic anti-inflammatory pathway and exhibits the inhibition of neuroinflammation in chronic pain. Trifluoro-icaritin (ICTF), a derivative of icaritin from the extract of a genus of Epimedium plant, is identified to possess profound anti-inflammatory activity. However, whether ICTF has anti-nociceptive effect on inflammatory pain and its potential mechanisms remain poorly elucidated. Intraperitoneal injection (i.p.) of ICTF to complete Freund's adjuvant (CFA)-induced inflammatory pain rats once daily for 21 consecutive days. Pain-related behaviors were evaluated with paw withdrawal threshold (PWT), paw withdrawal latency (PWL), and CatWalk gait analysis. Expression of pain-related signaling molecules in the spinal cord were detected using qRT-PCR, western blot assay, and immunofluorescence staining. This results showed that ICTF (3.0mg/kg, i.p.) effectively alleviated mechanical allodynia and thermal hyperalgesia not 0.3 and 1.0mg/kg in CFA rats. Subsequently, we further observed that ICTF (3.0mg/kg) dramatically decreased the mRNA and protein levels of HMGB1, NF-κB p65, and IL-1β but markedly enhanced α7nAChR and IL-10 expression in the spinal cord of CFA rats, and Immunofluorescence staining also showed that ICTF (3.0mg/kg) significantly increased the expression of α7nAChR and reduced IBA1 in the spinal cord of CFA rats, along with suppressing the alterations of gait parameters induced by CFA. Moreover, Intrathecal injection (i.t.) of α7nAChR antagonist alpha-bungarotoxin (α-Bgtx, 1.0μg/kg) not only reversed the anti-nociceptive effect of ICTF on pain hypersensitivity, but also inhibited the down-regulation of HMGB1, NF-κB p65, and IL-1β as well as the up-regulation of α7nAChR and IL-10 protein expression induced by ICTF treatment. Altogether, our results illustrate that ICTF enables to ameliorate CFA-induced inflammatory pain through α7nAChR-mediated inhibition of HMGB1/NF-κB signaling pathway in the spinal cord of rats, suggesting that ICTF may be exploited as a potential painkiller against chronic inflammatory pain.
Collapse
|
32
|
Ikeda T, Nakamura K, Kida T, Oku H. Possible roles of anti-type II collagen antibody and innate immunity in the development and progression of diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2022; 260:387-403. [PMID: 34379187 PMCID: PMC8786754 DOI: 10.1007/s00417-021-05342-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 11/08/2022] Open
Abstract
The pathogenesis of both diabetic retinopathy (DR) and rheumatoid arthritis (RA) has recently been considered to involve autoimmunity. Serum and synovial fluid levels of anti-type II collagen antibodies increase early after the onset of RA, thus inducing immune responses and subsequent hydrarthrosis and angiogenesis, which resemble diabetic macular edema and proliferative DR (PDR), respectively. We previously reported that DR is also associated with increased serum levels of anti-type II collagen antibodies. Retinal hypoxia in DR may induce pericytes to express type II collagen, resulting in autoantibody production against type II collagen. As the result of blood-retinal barrier disruption, anti-type II collagen antibodies in the serum come into contact with type II collagen around the retinal vessels. A continued loss of pericytes and type II collagen around the retinal vessels may result in a shift of the immune reaction site from the retina to the vitreous. It has been reported that anti-inflammatory M2 macrophages increased in the vitreous of PDR patients, accompanied by the activation of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a key regulator of innate immunity. M2 macrophages promote angiogenesis and fibrosis, which might be exacerbated and prolonged by dysregulated innate immunity.
Collapse
Affiliation(s)
- Tsunehiko Ikeda
- Department of Ophthalmology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan.
- Department of Ophthalmology, Osaka Kaisei Hospital, 1-6-10 Miyahara Yodogawa-ku, Osaka City, Osaka, Japan.
| | | | - Teruyo Kida
- Department of Ophthalmology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Hidehiro Oku
- Department of Ophthalmology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| |
Collapse
|
33
|
Singh TP, Carvalho AM, Sacramento LA, Grice EA, Scott P. Microbiota instruct IL-17A-producing innate lymphoid cells to promote skin inflammation in cutaneous leishmaniasis. PLoS Pathog 2021; 17:e1009693. [PMID: 34699567 PMCID: PMC8570469 DOI: 10.1371/journal.ppat.1009693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/05/2021] [Accepted: 10/12/2021] [Indexed: 12/26/2022] Open
Abstract
Innate lymphoid cells (ILCs) comprise a heterogeneous population of immune cells that maintain barrier function and can initiate a protective or pathological immune response upon infection. Here we show the involvement of IL-17A-producing ILCs in microbiota-driven immunopathology in cutaneous leishmaniasis. IL-17A-producing ILCs were RORγt+ and were enriched in Leishmania major infected skin, and topical colonization with Staphylococcus epidermidis before L. major infection exacerbated the skin inflammatory responses and IL-17A-producing RORγt+ ILC accumulation without impacting type 1 immune responses. IL-17A responses in ILCs were directed by Batf3 dependent CD103+ dendritic cells and IL-23. Moreover, experiments using Rag1-/- mice established that IL-17A+ ILCs were sufficient in driving the inflammatory responses as depletion of ILCs or neutralization of IL-17A diminished the microbiota mediated immunopathology. Taken together, this study indicates that the skin microbiota promotes RORγt+ IL-17A-producing ILCs, which augment the skin inflammation in cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Tej Pratap Singh
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (TPS); (PS)
| | - Augusto M. Carvalho
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Laís Amorim Sacramento
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elizabeth A. Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Phillip Scott
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (TPS); (PS)
| |
Collapse
|
34
|
Lopes ME, dos Santos LM, Sacks D, Vieira LQ, Carneiro MB. Resistance Against Leishmania major Infection Depends on Microbiota-Guided Macrophage Activation. Front Immunol 2021; 12:730437. [PMID: 34745100 PMCID: PMC8564857 DOI: 10.3389/fimmu.2021.730437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Innate immune cells present a dual role during leishmaniasis: they constitute the first line of host defense but are also the main host cells for the parasite. Response against the infection that results in the control of parasite growth and lesion healing depends on activation of macrophages into a classical activated phenotype. We report an essential role for the microbiota in driving macrophage and monocyte-derived macrophage activation towards a resistance phenotype against Leishmania major infection in mice. Both germ-free and dysbiotic mice showed a higher number of myeloid innate cells in lesions and increased number of infected cells, mainly dermal resident and inflammatory macrophages. Despite developing a Th1 immune response characterized by the same levels of IFN-γ production as the conventional mice, germ-free mice presented reduced numbers of iNOS+ macrophages at the peak of infection. Absence or disturbance of host microbiota impaired the capacity of bone marrow-derived macrophage to be activated for Leishmania killing in vitro, even when stimulated by Th1 cytokines. These cells presented reduced expression of inos mRNA, and diminished production of microbicidal molecules, such as ROS, while presenting a permissive activation status, characterized by increased expression of arginase I and il-10 mRNA and higher arginase activity. Colonization of germ-free mice with complete microbiota from conventional mice rescued their ability to control the infection. This study demonstrates the essential role of host microbiota on innate immune response against L. major infection, driving host macrophages to a resistance phenotype.
Collapse
Affiliation(s)
- Mateus Eustáquio Lopes
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Liliane Martins dos Santos
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Leda Quercia Vieira
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus B. Carneiro
- Laboratório de Gnotobiologia e Imunologia, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
35
|
Carneiro MB, Peters NC. The Paradox of a Phagosomal Lifestyle: How Innate Host Cell- Leishmania amazonensis Interactions Lead to a Progressive Chronic Disease. Front Immunol 2021; 12:728848. [PMID: 34557194 PMCID: PMC8452962 DOI: 10.3389/fimmu.2021.728848] [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: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Collapse
Affiliation(s)
- Matheus B Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
36
|
Fernández OL, Ramírez LG, Díaz-Varela M, Tacchini-Cottier F, Saravia NG. Neutrophil Activation: Influence of Antimony Tolerant and Susceptible Clinical Strains of L. (V.) panamensis and Meglumine Antimoniate. Front Cell Infect Microbiol 2021; 11:710006. [PMID: 34631596 PMCID: PMC8493214 DOI: 10.3389/fcimb.2021.710006] [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: 05/14/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Emerging evidence indicates that innate host response contributes to the therapeutic effect of antimicrobial medications. Recent studies have shown that Leishmania parasites derived by in vitro selection for resistance to pentavalent antimony (SbV) as meglumine antimoniate (MA) modulate the activation of neutrophils. However, whether modulation of neutrophil activation extends to natural resistance to this antileishmanial drug has not been established. We have evaluated the influence of clinical strains of L. (V.) panamensis having intrinsic tolerance/resistance to SbV, on the inflammatory response of neutrophils during ex vivo exposure to MA. Accordingly, neutrophils obtained from healthy donors were infected with clinical strains that are sensitive (n = 10) or intrinsically tolerant/resistant to SbV (n = 10) and exposed to a concentration approximating the maximal plasma concentration (Cmax) of SbV (32 µg/ml). The activation profile of neutrophils was evaluated as the expression of the surface membrane markers CD66b, CD18, and CD62L by flow cytometry, measurement of reactive oxygen species (ROS) by luminometry, and NET formation using Picogreen to measure dsDNA release and quantification of NETs by confocal microscopy. These parameters of activation were analyzed in relation with parasite susceptibility to SbV and exposure to MA. Here, we show that clinical strains presenting intrinsic tolerance/resistance to SbV induced significantly lower ROS production compared to drug-sensitive clinical strains, both in the presence and in the absence of MA. Likewise, analyses of surface membrane activation markers revealed significantly higher expression of CD62L on cells infected with intrinsically SbV tolerant/resistant L. (V.) panamensis than cells infected with drug-sensitive strains. Expression of other activation markers (CD18 and CD66b) and NET formation were similar for neutrophils infected with SbV sensitive and tolerant clinical strains under the conditions evaluated. Exposure to MA broadly impacted the activation of neutrophils, diminishing NET formation and the expression of CD62L, while augmenting ROS production and CD66b expression, independently of the parasite susceptibility phenotype. These results demonstrated that activation of human neutrophils ex vivo is differentially modulated by infection with clinical strains of L. (V.) panamensis having intrinsic tolerance/resistance to SbV compared to sensitive strains, and by exposure to antimonial drug.
Collapse
Affiliation(s)
- Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Lady Giovanna Ramírez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Míriam Díaz-Varela
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| |
Collapse
|
37
|
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: 14] [Impact Index Per Article: 4.7] [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
|
38
|
Nascimento MT, Franca M, Carvalho AM, Amorim CF, Peixoto F, Beiting D, Scott P, Carvalho EM, Carvalho LP. Inhibition of gamma-secretase activity without interfering in Notch signalling decreases inflammatory response in patients with cutaneous leishmaniasis. Emerg Microbes Infect 2021; 10:1219-1226. [PMID: 34009107 PMCID: PMC8676695 DOI: 10.1080/22221751.2021.1932608] [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] [Indexed: 11/18/2022]
Abstract
Cutaneous leishmaniasis (CL) patients present an exacerbated inflammatory response associated with tissue damage and ulcer development. Increasing numbers of patients have exhibited treatment failure, which remains not well understood. We hypothesized that adjuvant anti-inflammatory therapy would benefit CL patients. The aim of the present study was to investigate the contribution of Notch signalling and gamma-secretase activity to the inflammatory response observed in CL patients. Notch signalling is a molecular signalling pathway conserved among animal species. Gamma-secretase forms a complex of proteins that, among other pathways, modulates Notch signalling and immune response. We found that Notch 1 cell receptor signalling protects against the pathologic inflammatory response, and JLK6, a gamma-secretase inhibitor that does not interfere with Notch signalling, was shown to decrease the in-vitro inflammatory response in CL. Our data suggest that JLK6 may serve as an adjuvant treatment for CL patients.
Collapse
Affiliation(s)
- Maurício T Nascimento
- Laboratório de Pesquisas Clínicas; Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Mônica Franca
- Instituto de Ciências e Saúde Universidade Federal da Bahia, Salvador, Brazil
| | - Augusto M Carvalho
- Laboratório de Pesquisas Clínicas; Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Camila F Amorim
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Fábio Peixoto
- Laboratório de Pesquisas Clínicas; Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Daniel Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Phillip Scott
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Edgar M Carvalho
- Laboratório de Pesquisas Clínicas; Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Lucas P Carvalho
- Laboratório de Pesquisas Clínicas; Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto de Ciências e Saúde Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| |
Collapse
|
39
|
Sangare M, Coulibaly YI, Huda N, Vidal S, Tariq S, Coulibaly ME, Coulibaly SY, Soumaoro L, Dicko I, Traore B, Sissoko IM, Traore SF, Faye O, Nutman TB, Valenzuela JG, Oliveira F, Doumbia S, Kamhawi S, Semnani RT. Individuals co-exposed to sand fly saliva and filarial parasites exhibit altered monocyte function. PLoS Negl Trop Dis 2021; 15:e0009448. [PMID: 34106920 PMCID: PMC8189443 DOI: 10.1371/journal.pntd.0009448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/04/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In Mali, cutaneous leishmaniasis (CL) and filariasis are co-endemic. Previous studies in animal models of infection have shown that sand fly saliva enhance infectivity of Leishmania parasites in naïve hosts while saliva-specific adaptive immune responses may protect against cutaneous and visceral leishmaniasis. In contrast, the human immune response to Phlebotomus duboscqi (Pd) saliva, the principal sand fly vector in Mali, was found to be dichotomously polarized with some individuals having a Th1-dominated response and others having a Th2-biased response. We hypothesized that co-infection with filarial parasites may be an underlying factor that modulates the immune response to Pd saliva in endemic regions. METHODOLOGY/PRINCIPAL FINDINGS To understand which cell types may be responsible for polarizing human responses to sand fly saliva, we investigated the effect of salivary glands (SG) of Pd on human monocytes. To this end, elutriated monocytes were cultured in vitro, alone, or with SG, microfilariae antigen (MF ag) of Brugia malayi, or LPS, a positive control. The mRNA expression of genes involved in inflammatory or regulatory responses was then measured as were cytokines and chemokines associated with these responses. Monocytes of individuals who were not exposed to sand fly bites (mainly North American controls) significantly upregulated the production of IL-6 and CCL4; cytokines that enhance leishmania parasite establishment, in response to SG from Pd or other vector species. This selective inflammatory response was lost in individuals that were exposed to sand fly bites which was not changed by co-infection with filarial parasites. Furthermore, infection with filarial parasites resulted in upregulation of CCL22, a type-2 associated chemokine, both at the mRNA levels and by its observed effect on the frequency of recruited monocytes. CONCLUSIONS/SIGNIFICANCE Together, our data suggest that SG or recombinant salivary proteins from Pd alter human monocyte function by upregulating selective inflammatory cytokines.
Collapse
Affiliation(s)
- Moussa Sangare
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
- * E-mail: (MS); (RTS)
| | - Yaya Ibrahim Coulibaly
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Dermatology Hospital of Bamako, Bamako, Mali
| | - Naureen Huda
- Department of Pediatrics, University of California, San Francisco, California, United States of America
| | - Silvia Vidal
- Institut Recerca H. Sant Pau C. Sant Quintí, Spain
| | - Sameha Tariq
- Laboratory of Parasitic Diseases, LPD, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michel Emmanuel Coulibaly
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Siaka Yamoussa Coulibaly
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Lamine Soumaoro
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Ilo Dicko
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Bourama Traore
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Dermatology Hospital of Bamako, Bamako, Mali
| | - Ibrahim Moussa Sissoko
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Sekou Fantamady Traore
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Ousmane Faye
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Dermatology Hospital of Bamako, Bamako, Mali
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, LPD, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, LMVR, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Fabiano Oliveira
- Vector Molecular Biology Section, LMVR, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Seydou Doumbia
- Mali International Center for Excellence in Research, Faculty of Medicine and Odonto-Stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Shaden Kamhawi
- Vector Molecular Biology Section, LMVR, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Roshanak Tolouei Semnani
- Autoimmunity and Translational Immunology, Precigen, Inc. A wholly owned subsidiary of Intrexon Corporation, Germantown, Maryland, United States of America
- * E-mail: (MS); (RTS)
| |
Collapse
|
40
|
Yadav S, Verma V, Dhanda RS, Khurana S, Yadav M. Latent Upregulation of Nlrp3, Nlrc4 and Aim2 Differentiates between Asymptomatic and Symptomatic Trichomonas vaginalis Infection. Immunol Invest 2021; 51:1127-1148. [PMID: 33866944 DOI: 10.1080/08820139.2021.1909062] [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: 10/21/2022]
Abstract
Trichomonas vaginalis is a parasitic protozoan that causes trichomoniasis. The involvement of NLRP3 inflammasome in trichomoniasis has been discussed in recent studies. The present study aimed to find out the involvement of Nlrp3, Nlrc4, and Aim2 in the BALB/c mouse model infected with symptomatic and asymptomatic isolates of T. vaginalis by quantitative real-time PCR and immunohistochemistry. Our results showed a significantly increased expression of Nlrp3 in the vaginal tissue of the symptomatic group on the 2nd dpi and 14th dpi in the asymptomatic group, respectively. The cervical tissue of asymptomatic groups expressed higher Nlrp3 on 14th dpi than the symptomatic group. The Nlrc4 was expressed on 14th dpi in the vaginal and cervical tissues of mice infected with asymptomatic group as compared to the symptomatic group. Aim2 expression in vaginal tissue was highest at early time points in both the infected groups as compared to controls. However, in cervical tissues, a significant increase of Aim2 expression was observed on 14th dpi in asymptomatic as compared to the symptomatic group. The significantly higher expression of caspase-1 and caspase-4 was observed in cervical tissues of the asymptomatic group on 14th dpi as compared to the symptomatic group, respectively. All NLRs together resulted in higher IL-1β expression in the vaginal tissues of the symptomatic and asymptomatic groups. We conclude from this study that early expression of Nlrp3, Nlrc4, and Aim2 was seen in the symptomatic group as compared to the late-onset asymptomatic in the vaginal and cervical tissues.
Collapse
Affiliation(s)
- Sonal Yadav
- Dr. B. R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | - Vivek Verma
- Dr. B. R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | | | - Sumeeta Khurana
- Department of Medical Parasitology, PGIMER, Chandigarh, India
| | - Manisha Yadav
- Dr. B. R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India
| |
Collapse
|
41
|
Bichiou H, Bouabid C, Rabhi I, Guizani-Tabbane L. Transcription Factors Interplay Orchestrates the Immune-Metabolic Response of Leishmania Infected Macrophages. Front Cell Infect Microbiol 2021; 11:660415. [PMID: 33898331 PMCID: PMC8058464 DOI: 10.3389/fcimb.2021.660415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Leishmaniasis is a group of heterogenous diseases considered as an important public health problem in several countries. This neglected disease is caused by over 20 parasite species of the protozoa belonging to the Leishmania genus and is spread by the bite of a female phlebotomine sandfly. Depending on the parasite specie and the immune status of the patient, leishmaniasis can present a wide spectrum of clinical manifestations. As an obligate intracellular parasite, Leishmania colonize phagocytic cells, mainly the macrophages that orchestrate the host immune response and determine the fate of the infection. Once inside macrophages, Leishmania triggers different signaling pathways that regulate the immune and metabolic response of the host cells. Various transcription factors regulate such immune-metabolic responses and the associated leishmanicidal and inflammatory reaction against the invading parasite. In this review, we will highlight the most important transcription factors involved in these responses, their interactions and their impact on the establishment and the progression of the immune response along with their effect on the physiopathology of the disease.
Collapse
Affiliation(s)
- Haifa Bichiou
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Biotechnology Department, Higher Institute of Biotechnology at Sidi-Thabet (ISBST), Biotechpole Sidi-Thabet- University of Manouba, Tunis, Tunisia
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
| |
Collapse
|
42
|
Novais FO, Amorim CF, Scott P. Host-Directed Therapies for Cutaneous Leishmaniasis. Front Immunol 2021; 12:660183. [PMID: 33841444 PMCID: PMC8032888 DOI: 10.3389/fimmu.2021.660183] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
Cutaneous leishmaniasis exhibits a wide spectrum of clinical presentations from self-resolving infections to severe chronic disease. Anti-parasitic drugs are often ineffective in the most severe forms of the disease, and in some cases the magnitude of the disease can result from an uncontrolled inflammatory response rather than unrestrained parasite replication. In these patients, host-directed therapies offer a novel approach to improve clinical outcome. Importantly, there are many anti-inflammatory drugs with known safety and efficacy profiles that are currently used for other inflammatory diseases and are readily available to be used for leishmaniasis. However, since leishmaniasis consists of a wide range of clinical entities, mediated by a diverse group of leishmanial species, host-directed therapies will need to be tailored for specific types of leishmaniasis. There is now substantial evidence that host-directed therapies are likely to be beneficial beyond autoimmune diseases and cancer and thus should be an important component in the armamentarium to modulate the severity of cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Fernanda O Novais
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Camila Farias Amorim
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
43
|
Passelli K, Billion O, Tacchini-Cottier F. The Impact of Neutrophil Recruitment to the Skin on the Pathology Induced by Leishmania Infection. Front Immunol 2021; 12:649348. [PMID: 33732265 PMCID: PMC7957080 DOI: 10.3389/fimmu.2021.649348] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/04/2021] [Indexed: 12/29/2022] Open
Abstract
Leishmania (L.) are obligate intracellular protozoan parasites that cause the leishmaniases, a spectrum of neglected infectious vector-borne diseases with a broad range of clinical manifestations ranging from local cutaneous, to visceral forms of the diseases. The parasites are deposited in the mammalian skin during the blood meal of an infected female phlebotomine sand fly. The skin is a complex organ acting as the first line of physical and immune defense against pathogens. Insults to skin integrity, such as that occurring during insect feeding, induces the local secretion of pro-inflammatory molecules generating the rapid recruitment of neutrophils. At the site of infection, skin keratinocytes play a first role in host defense contributing to the recruitment of inflammatory cells to the infected dermis, of which neutrophils are the first recruited cells. Although neutrophils efficiently kill various pathogens including Leishmania, several Leishmania species have developed mechanisms to survive in these cells. In addition, through their rapid release of cytokines, neutrophils modulate the skin microenvironment at the site of infection, a process shaping the subsequent development of the adaptive immune response. Neutrophils may also be recruited later on in unhealing forms of cutaneous leishmaniasis and to the spleen and liver in visceral forms of the disease. Here, we will review the mechanisms involved in neutrophil recruitment to the skin following Leishmania infection focusing on the role of keratinocytes in this process. We will also discuss the distinct involvement of neutrophils in the outcome of leishmaniasis.
Collapse
Affiliation(s)
- Katiuska Passelli
- Department of Biochemistry, WHO Collaborative Centre for Research and Training in Immunology, University of Lausanne, Lausanne, Switzerland
| | - Oaklyne Billion
- Department of Biochemistry, WHO Collaborative Centre for Research and Training in Immunology, University of Lausanne, Lausanne, Switzerland
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, WHO Collaborative Centre for Research and Training in Immunology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
44
|
Guerra JM, Fernandes NCCA, Réssio RA, Kimura LM, Barbosa JER, Taniguchi HH, Hiramoto RM, Motoie G, Tolezano JE, Cogliati B. Cytokine profile and parasite load in lymph nodes of dogs naturally infected with Leishmania infantum from distinct epidemiological scenarios in São Paulo State, Brazil. Vet Immunol Immunopathol 2021; 233:110198. [PMID: 33548792 DOI: 10.1016/j.vetimm.2021.110198] [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: 01/25/2020] [Revised: 01/14/2021] [Accepted: 01/23/2021] [Indexed: 10/22/2022]
Abstract
Visceral leishmaniasis (VL) is an important zoonotic vector-borne disease and domestic dogs are considered the main domiciliary and peri-domiciliary reservoir of Leishmania (Leishmania) infantum in South America. Distinct eco-epidemiological scenarios associated to the prevalence of the disease, clusters of parasite genotypes and chemotypes of vectors population are described in Brazil, especially in the state of São Paulo (SP). In this context, the purpose of the present study is to evaluate the clinical signs, histopathological lesions, parasite load and cytokine profile by immunohistochemistry (IHC) in popliteal lymph nodes of canines naturally infected with L. infantum, from different municipalities of the state of SP. Eighty-three dogs with VL, 61 from northwest SP (NWSP) and 22 from southeast SP (SESP), were clinically classified in stage II, with no babesiosis and ehrlichiosis. Subcapsular inflammatory infiltration and histiocytosis were significantly higher in the SESP group (p = 0.0128; 0.0077, respectively). On the other hand, dogs from NWSP revealed 4.6-fold significantly higher parasite burden (p = 0.0004) and higher IHC scores of IL-1β (p = 0.0275) and IL-4 (p = 0.0327) in the popliteal lymph node tissues, which may be associated with the susceptibility and progression of the disease in these dogs. Differences in immune response profile associated with higher parasite load in dogs can also contribute to explain the distinct eco-epidemiological patterns of VL in specific geographic regions.
Collapse
Affiliation(s)
- Juliana M Guerra
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil; Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, Av. Prof. Orlando Marques de Paiva, 87, São Paulo, SP, 05508-270, Brazil.
| | - Natália C C A Fernandes
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil; Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, Av. Prof. Orlando Marques de Paiva, 87, São Paulo, SP, 05508-270, Brazil
| | - Rodrigo A Réssio
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - Lidia M Kimura
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - José E R Barbosa
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - Helena H Taniguchi
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - Roberto M Hiramoto
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - Gabriela Motoie
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - José E Tolezano
- Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças, Secretaria do Estado daSaúde de São Paulo, Avenida Dr. Arnaldo, 351, 01246-000, São Paulo, SP, Brazil
| | - Bruno Cogliati
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, Av. Prof. Orlando Marques de Paiva, 87, São Paulo, SP, 05508-270, Brazil
| |
Collapse
|
45
|
Regulation of macrophage subsets and cytokine production in leishmaniasis. Cytokine 2020; 147:155309. [PMID: 33334669 DOI: 10.1016/j.cyto.2020.155309] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022]
Abstract
Macrophages are host cells for parasites of the genus Leishmania where they multiply inside parasitophorous vacuoles. Paradoxically, macrophages are also the cells responsible for killing or controlling parasite growth, if appropriately activated. In this review, we will cover the patterns of macrophage activation and the mechanisms used by the parasite to circumvent being killed. We will highlight the impacts of the vector bite on macrophage activation. Finally, we will discuss the ontogeny of macrophages that are infected by Leishmania spp.
Collapse
|
46
|
Osero BO, Aruleba RT, Brombacher F, Hurdayal R. Unravelling the unsolved paradoxes of cytokine families in host resistance and susceptibility to Leishmania infection. Cytokine X 2020; 2:100043. [PMID: 33415318 PMCID: PMC7773805 DOI: 10.1016/j.cytox.2020.100043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022] Open
Abstract
Leishmaniasis is a neglected disease caused by protozoan parasites of the genus Leishmania. Successful clearance of Leishmania relies on a robust human immune response and various cytokines have been implicated in resistance and susceptibility to Leishmania infection. Accordingly, various immunotherapeutic approaches involving cytokines and cytokine receptors are being considered as novel avenues of treatment given the limited efficacy of current anti-leishmanial drugs. These approaches target canonical T helper (Th)1/Type 1 cytokines as intended mediators of host-protection to infection whilst concomitantly suppressing Th2/Type 2 cytokines and their anticipated disease-promoting roles. However, the use of cytokine and cytokine receptor gene-deficient mice over the years has challenged this simplistic view of Th1/Type 1-mediated resistance and Th2/Type 2-mediated susceptibility. Indeed, contribution to susceptibility vs resistance is only a partial consequence to cytokine action as the overall response is multi-faceted due to the pleiotropic, redundant, antagonistic and synergistic action of cytokines and interactions with immune cells in the diseased state. Notably, while the responses of certain cytokines are selectively host-protective or characteristic disease-enhancers, some ligands exert a response depending on the parasite-species initiating infection. Paradoxically, others play dual or contradictory roles in different Leishmania immunopathologies. Hence, cytokines in disease is an unsolved paradox and a comprehensive knowledge of cytokine interplay is important to guide the development of novel immunotherapeutics against leishmaniasis. In this review, we characterize various cytokine families in persistence and clearance of the Leishmania parasite and particularly elucidate unsolved cytokine puzzles in leishmaniasis based on information acquired from "gain of knowledge by loss of function" studies in cytokine and cytokine receptor gene-deficient mice.
Collapse
Affiliation(s)
- Bernard Ong'ondo Osero
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Raphael Taiwo Aruleba
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Ramona Hurdayal
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Observatory 7925, Cape Town, South Africa
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, Cape Town, South Africa
| |
Collapse
|
47
|
Evaluation of a New Topical Treatment for the Control of Cutaneous Leishmaniasis. Microorganisms 2020; 8:microorganisms8111803. [PMID: 33212818 PMCID: PMC7696824 DOI: 10.3390/microorganisms8111803] [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: 09/01/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
Leishmania major (L. major) causes cutaneous leishmaniasis in the Old World. The infection mostly induces a localized lesion restricted to the sand fly bite. The costs and the side effects of current treatments render imperative the development of new therapies that are affordable and easy to administrate. Topical treatment would be the ideal option for the treatment of cutaneous leishmaniasis. MF29 is a 3-haloacetamidobenzoate that was shown in vitro to inhibit tubulin assembly in Leishmania. Here, we tested a topical cream formulated with MF29. BALB/c mice were infected in the ear dermis with L. major metacyclic promastigotes and once the lesion appeared, mice were treated with different concentrations of MF29 and compared to the control group treated with the cream used as the vehicle. We observed that topical application of MF29 reduced the progression of the infection while control groups developed an unhealing lesion that became necrotic. The treatment decreased the type 2 immune response. Comparison with SinaAmphoLeish, another topical treatment, revealed that MF29 treatment once a day was sufficient to control lesion development, while application SinaAmphoLeish needed applications twice daily. Collectively, our data suggest that MF-29 topical application could be a promising topical treatment for cutaneous leishmaniasis.
Collapse
|
48
|
Bhattacharya P, Dey R, Saxena A, Karmakar S, Ismail N, Gannavaram S, Dagur PK, Satoskar M, Satoskar S, De Paoli S, Takeda K, McCoy JP, Nakhasi HL. Essential Role of Neutrophils in the Protective Immune Response Induced by a Live Attenuated Leishmania Vaccine. THE JOURNAL OF IMMUNOLOGY 2020; 205:3333-3347. [PMID: 33177159 DOI: 10.4049/jimmunol.2000829] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
No licensed vaccine exists against visceral leishmaniasis (VL), a disease caused by the Leishmania donovani parasite. We have previously reported both macrophages and dendritic cells play important role in the protection induced by a live attenuated centrin gene-deleted L. donovani (LdCen-/- ) parasite vaccine. The role of neutrophils in orchestrating the initial innate response to pathogens is widely recognized. To investigate the early interaction of LdCen-/- with neutrophils, we immunized mice intradermally in the ear pinna with LdCen-/- Compared with LdWT infection, LdCen-/- parasites induced higher recruitment of neutrophils to the ear dermis and ear draining lymph nodes (dLN) as early as 6-18 h after immunization, which were predominantly proinflammatory in nature. Neutrophils from ear dLN of LdCen-/- -immunized mice exhibited heightened expression of costimulatory molecules and attenuated expression of coinhibitory molecules necessary for higher T cell activation. Further phenotypic characterization revealed heterogeneous neutrophil populations containing Nα and Nβ subtypes in the ear dLN. Of the two, the parasitized Nα subset from LdCen-/- -immunized mice exhibited much stronger Ag-specific CD4+ T cell proliferation ex vivo. Adoptive transfer of neutrophils bearing LdCen-/- parasites induced an increased Th1 response in naive mice. Importantly, neutrophil depletion significantly abrogated Ag-specific CD4+ T cell proliferation in LdCen-/- -immunized mice and impaired protection against virulent challenge. Conversely, replenishing of neutrophils significantly restored the LdCen-/- -induced host-protective response. These results suggest that neutrophils are indispensable for protective immunity induced by LdCen-/- parasite vaccine.
Collapse
Affiliation(s)
- Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Ankit Saxena
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | - Silvia De Paoli
- Office of Blood Research and Review, U.S. Food and Drug Administration, Silver Spring, MD 20993; and
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - John Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| |
Collapse
|
49
|
Chaves MM, Lee SH, Kamenyeva O, Ghosh K, Peters NC, Sacks D. The role of dermis resident macrophages and their interaction with neutrophils in the early establishment of Leishmania major infection transmitted by sand fly bite. PLoS Pathog 2020; 16:e1008674. [PMID: 33137149 PMCID: PMC7660907 DOI: 10.1371/journal.ppat.1008674] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/12/2020] [Accepted: 09/24/2020] [Indexed: 11/18/2022] Open
Abstract
There is substantial experimental evidence to indicate that Leishmania infections that are transmitted naturally by the bites of infected sand flies differ in fundamental ways from those initiated by needle inocula. We have used flow cytometry and intravital microscopy (IVM) to reveal the heterogeneity of sand fly transmission sites with respect to the subsets of phagocytes in the skin that harbor L. major within the first hours and days after infection. By flow cytometry analysis, dermis resident macrophages (TRMs) were on average the predominant infected cell type at 1 hr and 24 hr. By confocal IVM, the co-localization of L. major and neutrophils varied depending on the proximity of deposited parasites to the presumed site of vascular damage, defined by the highly localized swarming of neutrophils. Some of the dermal TRMs could be visualized acquiring their infections via transfer from or efferocytosis of parasitized neutrophils, providing direct evidence for the "Trojan Horse" model. The role of neutrophil engulfment by dermal TRMs and the involvement of the Tyro3/Axl/Mertk family of receptor tyrosine kinases in these interactions and in sustaining the anti-inflammatory program of dermal TRMs was supported by the effects observed in neutrophil depleted and in Axl-/-Mertk-/- mice. The Axl-/-Mertk-/- mice also displayed reduced parasite burdens but more severe pathology following L. major infection transmitted by sand fly bite.
Collapse
Affiliation(s)
- Mariana M. Chaves
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, Unites States of America
| | - Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, Unites States of America
- * E-mail: (SHL); (DS)
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, Unites States of America
| | - Kashinath Ghosh
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, Unites States of America
| | - Nathan C. Peters
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, Unites States of America
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, Unites States of America
- * E-mail: (SHL); (DS)
| |
Collapse
|
50
|
Involvement of the Inflammasome and Th17 Cells in Skin Lesions of Human Cutaneous Leishmaniasis Caused by Leishmania ( Viannia) panamensis. Mediators Inflamm 2020; 2020:9278931. [PMID: 33192178 PMCID: PMC7641710 DOI: 10.1155/2020/9278931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/24/2020] [Accepted: 09/19/2020] [Indexed: 01/07/2023] Open
Abstract
Localized cutaneous leishmaniasis (LCL) caused by Leishmania (Viannia) panamensis is an endemic disease in Panama. This condition causes ulcerated skin lesions characterized by a mixed Th1/Th2 immune response that is responsible for disease pathology. However, the maintenance of the in situ inflammatory process involves other elements, such as Th17 and inflammasome responses. Although these processes are associated with parasite elimination, their role in the increase in disease pathology cannot be discarded. Thus, the role in Leishmania infection is still unclear. In this sense, the present study aimed at characterizing the Th17 and inflammasome responses in the skin lesions of patients with LCL caused by L. (V.) panamensis to help elucidate the pathogenesis of this disease in Panama. Th17 and inflammasome responses were evaluated by immunohistochemistry (IHQ) in 46 skin biopsies from patients with LCL caused by L. (V.) panamensis. The Th17 immune response was assessed using CD3, CD4, RoRγt, IL-17, IL-6, IL-23, and TGF-β1 antibodies, and the inflammasome response was assessed by IL-1β, IL-18, and caspase-1 antibodies. The presence of the Th17 and inflammasome responses was evidenced by a positive reaction for all immunological markers in the skin lesions. An inverse correlation between the density of amastigotes and the density of RoRγt+, IL-17+, IL-1β+, and caspase-1+ cells was observed, but no correlation between Th17 and the inflammasome response with evolutionary disease pathology was reported. These data showed the participation of Th17 cells and the inflammasome in the inflammatory response of the skin lesions of LCL caused by L. (V.) panamensis infection. These results suggest a role in the control of tissue parasitism of IL-17 and the activation of the NLRP3 inflammasome dependent on IL-1β but cannot exclude their role in the development of disease pathology.
Collapse
|