1
|
Ahmad F, Ahmad S, Srivastav AK, Upadhyay TK, Husain A, Khubaib M, Kang S, Park MN, Kim B, Sharma R. "β-glucan signalling stimulates NOX-2 dependent autophagy and LC-3 associated autophagy (LAP) pathway". Int J Biol Macromol 2024; 282:136520. [PMID: 39401634 DOI: 10.1016/j.ijbiomac.2024.136520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 12/19/2024]
Abstract
β-Glucan, a complex polysaccharide derived from fungal and yeast cell walls, plays a crucial role in modulating immune responses through their interaction with receptors such as Dectin-1 and Complement receptor 3 (CR-3). This review provides an in-depth analysis of the molecular mechanisms by which β-glucans activate receptor-mediated signalling pathways, focusing particularly on the LC3-associated phagocytosis (LAP) and autophagy pathways. Hence, we explore how β-glucan receptor engagement stimulates NADPH oxidase 2 (NOX-2), leading to the intracellular production of significant level of reactive oxygen species (ROS) essential for both conventional autophagy and LAP. While significant progress has been made in elucidation of downstream signaling by glucans, the regulation of phago-lysosomal maturation and antigen presentation during LAP induction still remains less explored. This review aims to provide a comprehensive overview of these pathways and their regulation by β-glucans. By consolidating the current knowledge, we seek to highlight how these mechanisms can be leveraged for therapeutic applications, particularly in the context of tuberculosis (TB) management, where β-glucans could serve as host-directed adjuvant therapies to combat drug-resistant strains. Despite major advancements in this field, currently key research gaps still persist, including detailed molecular interactions between β-glucan receptors and NOX-2 and the translation of these findings to in-vivo models and clinical investigations. This review underscores the need for further research to explore the therapeutic potential of β-glucans in managing not only tuberculosis but also other diseases such as cancer, cardiovascular conditions, and metabolic disorders.
Collapse
Affiliation(s)
- Firoz Ahmad
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India; Department of Physiological Sciences, Oklahoma Centre for Respiratory and Infectious Diseases, Oklahoma State University, OK 74074, United States of America
| | - Shad Ahmad
- Department of Biochemistry, Dr. Ram Manohar Lohia Avadh University, Faizabad 224001, Uttar Pradesh, India
| | - Anurag Kumar Srivastav
- Department of Clinical Immunology & Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Tarun Kumar Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences & Research and Development Cell, Parul University, Vadodara 391760, Gujarat, India
| | - Adil Husain
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India; Department of Biochemistry, Babu Banarasi Das [BBD] College of Dental Sciences BBD University, Lucknow 226028, Uttar Pradesh, India
| | - Mohd Khubaib
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea.
| | - Rolee Sharma
- Department of Life Sciences & Biotechnology, CSJM University, Kanpur 228024, Uttar Pradesh, India.
| |
Collapse
|
2
|
Arfath Y, Kotra T, Faizan MI, Akhtar A, Abdullah ST, Ahmad T, Ahmed Z, Rayees S. TRPV4 facilitates the reprogramming of inflamed macrophages by regulating IL-10 production via CREB. Inflamm Res 2024; 73:1687-1697. [PMID: 39101955 DOI: 10.1007/s00011-024-01923-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Transient receptor potential vanilloid type 4 (TRPV4) is a versatile ion channel with diverse roles in immune cells, including macrophages. While its function in inflammation remains debated, we investigated its role in regulating IL-10 production and its impact on macrophage reprogramming during inflammation. METHODS We investigated the connection between TRPV4 activation and CREB-mediated IL-10 production during inflammation. Notably, this signaling pathway was found to reprogram macrophages and enhance their ability to resist inflammatory damage. The experiments were conducted on primary macrophages and were further corroborated by animal studies. RESULTS In response to TRPV4 activation during inflammation, we observed a significant increase in intracellular Ca2+ levels, which triggered the activation of the transcription factor CREB, subsequently upregulating IL-10 production. This IL-10 played a pivotal role in reprogramming macrophages to withstand inflammatory damage. Using a mouse model of acute lung injury (ALI), we confirmed that TRPV4 activation during ALI led to IL-10 secretion, but this increase did not significantly contribute to inflammation resolution. Moreover, we found that TRPV4 prevented the accumulation of dysfunctional mitochondria in macrophages through the CREB-IL-10 axis during inflammation. Suppression of CREB or TRPV4 inhibition exacerbated mitochondrial dysfunction, while treatment with recombinant IL-10 mitigated these effects. Additionally, IL-10 induced mitophagy and cleared dysfunctional mitochondria in LPS-exposed cells. CONCLUSION Our study highlights the essential role of TRPV4 in regulating IL-10 production and mitochondrial health in macrophages during inflammation. These findings contribute to understand the role of TRPV4 in immune responses and suggest potential therapeutic targets for modulating inflammation-induced cellular dysfunction.
Collapse
Affiliation(s)
- Yassir Arfath
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tusharika Kotra
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Md Imam Faizan
- Multidisciplinary Centre for Advanced Research and Studies, JMI, New Delhi, 110025, India
| | - Areej Akhtar
- Multidisciplinary Centre for Advanced Research and Studies, JMI, New Delhi, 110025, India
| | - Sheikh Tasduq Abdullah
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, JMI, New Delhi, 110025, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
| | - Sheikh Rayees
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
3
|
Almeida P, Fernandes Â, Alves I, Pinho SS. "Glycans in Trained Immunity: Educators of innate immune memory in homeostasis and disease". Carbohydr Res 2024; 544:109245. [PMID: 39208605 DOI: 10.1016/j.carres.2024.109245] [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: 06/21/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Trained Immunity is defined as a biological process normally induced by exogenous or endogenous insults that triggers epigenetic and metabolic reprogramming events associated with long-term adaptation of innate immune cells. This trained phenotype confers enhanced responsiveness to subsequent triggers, resulting in an innate immune "memory" effect. Trained Immunity, in the past decade, has revealed important benefits for host defense and homeostasis, but can also induce potentially harmful outcomes associated with chronic inflammatory disorders or autoimmune diseases. Interestingly, evidence suggest that the "trainers" prompting trained immunity are frequently glycans structures. In fact, the exposure of different types of glycans at the surface of pathogens is a key driver of the training phenotype, leading to the reprogramming of innate immune cells through the recognition of those glycan-triggers by a variety of glycan-binding proteins (GBPs) expressed by the immune cells. β-glucan or mannose-enriched structures in Candida albicans are some of the examples that highlight the potential of glycans in trained immunity, both in homeostasis and in disease. In this review, we will discuss the relevance of glycans exposed by pathogens in establishing key immunological hubs with glycan-recognizing receptors expressed in immune cells, highlighting how this glycan-GBP network can impact trained immunity. Finally, we discuss the power of glycans and GBPs as potential targets in trained immunity, envisioning potential therapeutic applications.
Collapse
Affiliation(s)
- Pedro Almeida
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
| | - Ângela Fernandes
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
| | - Inês Alves
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
| | - Salomé S Pinho
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal; Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
4
|
Lensch V, Gabba A, Hincapie R, Bhagchandani SH, Basak A, Alam MM, Noble J, Irvine DJ, Shalek AK, Johnson JA, Finn MG, Kiessling LL. Carbohydrate-Lectin Interactions Reprogram Dendritic Cells to Promote Type 1 Anti-Tumor Immunity. ACS NANO 2024; 18:26770-26783. [PMID: 39283240 PMCID: PMC11646345 DOI: 10.1021/acsnano.4c07360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Cancer vaccine development is inhibited by a lack of strategies for directing dendritic cell (DC) induction of effective tumor-specific cellular immunity. Pathogen engagement of DC lectins and toll-like receptors (TLRs) is thought to shape immunity by directing T cell function. Controlling downstream responses, however, remains a major challenge. A critical goal in advancing vaccine development involves the identification of receptors that drive type 1 cellular immunity. The immune system monitors cells for aberrant glycosylation (a sign of a foreign entity), but potent activation occurs when a second signal, such as single-stranded RNA or lipopolysaccharide, is present to activate TLR signaling. To exploit dual signaling, we engineered a glycan-costumed virus-like particle (VLP) vaccine that displays a DC-SIGN-selective aryl mannose ligand and encapsulates TLR7 agonists. These VLPs deliver programmable peptide antigens to induce robust DC activation and type 1 cellular immunity. In contrast, VLPs lacking this critical DC-SIGN ligand promoted DC-mediated humoral immunity, offering limited tumor control. Vaccination with glycan-costumed VLPs generated tumor antigen-specific Th1 CD4+ and CD8+ T cells that infiltrated solid tumors, significantly inhibiting tumor growth in a murine melanoma model. The tailored VLPs also afforded protection against the reintroduction of tumor cells. Thus, DC lectin-driven immune reprogramming, combined with the modular programmability of VLP platforms, provides a promising framework for directing cellular immunity to advance cancer immunotherapies and vaccines.
Collapse
Affiliation(s)
- Valerie Lensch
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Adele Gabba
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Sachin H Bhagchandani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ankit Basak
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Mohammad Murshid Alam
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jeffery Noble
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| | - Alex K Shalek
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - M G Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
5
|
Santisteban Celis IC, Matoba N. Lectibodies as antivirals. Antiviral Res 2024; 227:105901. [PMID: 38734211 DOI: 10.1016/j.antiviral.2024.105901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Growing concerns regarding the emergence of highly transmissible viral diseases highlight the urgent need to expand the repertoire of antiviral therapeutics. For this reason, new strategies for neutralizing and inhibiting these viruses are necessary. A promising approach involves targeting the glycans present on the surfaces of enveloped viruses. Lectins, known for their ability to recognize specific carbohydrate molecules, offer the potential for glycan-targeted antiviral strategies. Indeed, numerous studies have reported the antiviral effects of various lectins of both endogenous and exogenous origins. However, many lectins in their natural forms, are not suitable for use as antiviral therapeutics due to toxicity, other unfavorable pharmacological effects, and/or unreliable manufacturing sources. Therefore, improvements are crucial for employing lectins as effective antiviral therapeutics. A novel approach to enhance lectins' suitability as pharmaceuticals could be the generation of recombinant lectin-Fc fusion proteins, termed "lectibodies." In this review, we discuss the scientific rationale behind lectin-based antiviral strategies and explore how lectibodies could facilitate the development of new antiviral therapeutics. We will also share our perspective on the potential of these molecules to transcend their potential use as antiviral agents.
Collapse
Affiliation(s)
- Ian Carlosalberto Santisteban Celis
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville School of Medicine, Louisville, KY, USA
| | - Nobuyuki Matoba
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville School of Medicine, Louisville, KY, USA; UofL Health - Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
6
|
Lefèbre J, Falk T, Ning Y, Rademacher C. Secondary Sites of the C-type Lectin-Like Fold. Chemistry 2024; 30:e202400660. [PMID: 38527187 DOI: 10.1002/chem.202400660] [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: 02/18/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
C-type lectins are a large superfamily of proteins involved in a multitude of biological processes. In particular, their involvement in immunity and homeostasis has rendered them attractive targets for diverse therapeutic interventions. They share a characteristic C-type lectin-like domain whose adaptability enables them to bind a broad spectrum of ligands beyond the originally defined canonical Ca2+-dependent carbohydrate binding. Together with variable domain architecture and high-level conformational plasticity, this enables C-type lectins to meet diverse functional demands. Secondary sites provide another layer of regulation and are often intricately linked to functional diversity. Located remote from the canonical primary binding site, secondary sites can accommodate ligands with other physicochemical properties and alter protein dynamics, thus enhancing selectivity and enabling fine-tuning of the biological response. In this review, we outline the structural determinants allowing C-type lectins to perform a large variety of tasks and to accommodate the ligands associated with it. Using the six well-characterized Ca2+-dependent and Ca2+-independent C-type lectin receptors DC-SIGN, langerin, MGL, dectin-1, CLEC-2 and NKG2D as examples, we focus on the characteristics of non-canonical interactions and secondary sites and their potential use in drug discovery endeavors.
Collapse
Affiliation(s)
- Jonathan Lefèbre
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Torben Falk
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Yunzhan Ning
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| |
Collapse
|
7
|
Borborema MEDA, Miranda DEDO, de Lucena TMC, de Lorena VMB, Rabello MCDS, de Azevêdo Silva J. Steroid immune responsive gene regulation in Mycobacterium tuberculosis infection in vitro. Tuberculosis (Edinb) 2024; 146:102497. [PMID: 38408402 DOI: 10.1016/j.tube.2024.102497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 02/28/2024]
Abstract
Tuberculosis (TB) is an infectious disease displaying a multifactorial pathology. The immunomodulatory role attributed to steroid hormones, such as vitamin D3 (VD3) and 17β-estradiol (E2), highlighted the importance of these hormones against Mycobacterium tuberculosis (Mtb) infection. In order to understand their influence upon gene expression of immune and inflammatory responsive genes against Mtb we tested it in vitro using peripheral blood mononuclear cells (PBMCs). Cells were pretreated with VD3 (50 ng/mL) or E2 (100 nM/mL) and co-cultured with H37Rv Mtb or stimulated with lipopolysaccharide from Escherichia coli (LPS). After 24 h and 72 h of co-culture the Mtb viability in macrophages test was performed, as well the total RNA isolation for gene expression analysis by RT-qPCR of the following target genes: NLRP3, DC-SIGN, IL-1β, and IL-10. We also measured IL-10, TNF, IFN-γ, IL-4, IL-6, and IL-2 supernatant levels. As the main results, we found that VD3 and E2 downregulated the expression of inflammatory genes NLRP3, IL-1β, and IL-10 expression in Mtb co-cultured cells. Finally, VD3 treatment increased the release of the cytokine IFN-γ in Mtb-infected cells, while E2 treatment inhibited the release of IL-10, TNF, IFN-γ, and IL-6. Therefore, we report an immunogenetic influence of VD3 and E2 upon Mtb co-culture.
Collapse
Affiliation(s)
- Maria Eduarda de Albuquerque Borborema
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Débora Elienai de Oliveira Miranda
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Thays Maria Costa de Lucena
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - Jaqueline de Azevêdo Silva
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
8
|
Pither MD, Andretta E, Rocca G, Balzarini F, Matamoros-Recio A, Colicchio R, Salvatore P, van Kooyk Y, Silipo A, Granucci F, Martin-Santamaria S, Chiodo F, Molinaro A, Di Lorenzo F. Deciphering the Chemical Language of the Immunomodulatory Properties of Veillonella parvula Lipopolysaccharide. Angew Chem Int Ed Engl 2024; 63:e202401541. [PMID: 38393988 DOI: 10.1002/anie.202401541] [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: 01/22/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 02/25/2024]
Abstract
Veillonella parvula, prototypical member of the oral and gut microbiota, is at times commensal yet also potentially pathogenic. The definition of the molecular basis tailoring this contrasting behavior is key for broadening our understanding of the microbiota-driven pathogenic and/or tolerogenic mechanisms that take place within our body. In this study, we focused on the chemistry of the main constituent of the outer membrane of V. parvula, the lipopolysaccharide (LPS). LPS molecules indeed elicit pro-inflammatory and immunomodulatory responses depending on their chemical structures. Herein we report the structural elucidation of the LPS from two strains of V. parvula and show important and unprecedented differences in both the lipid and carbohydrate moieties, including the identification of a novel galactofuranose and mannitol-containing O-antigen repeating unit for one of the two strains. Furthermore, by harnessing computational studies, in vitro human cell models, as well as lectin binding solid-phase assays, we discovered that the two chemically diverse LPS immunologically behave differently and have attempted to identify the molecular determinant(s) governing this phenomenon. Whereas pro-inflammatory potential has been evidenced for the lipid A moiety, by contrast a plausible "immune modulating" action has been proposed for the peculiar O-antigen portion.
Collapse
Affiliation(s)
- Molly Dorothy Pither
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 4, 80126, Naples, Italy
| | - Emanuela Andretta
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 4, 80126, Naples, Italy
| | - Giuseppe Rocca
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1, 20126, Milan, Italy
| | - Fabio Balzarini
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB, Amsterdam, The Netherlands
| | - Alejandra Matamoros-Recio
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, CIB-CSIC, C/ Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB, Amsterdam, The Netherlands
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 4, 80126, Naples, Italy
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1, 20126, Milan, Italy
| | - Sonsoles Martin-Santamaria
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, CIB-CSIC, C/ Ramiro de Maeztu, 9, 28040, Madrid, Spain
| | - Fabrizio Chiodo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB, Amsterdam, The Netherlands
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei, 34, 80078, Pozzuoli, Naples, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 4, 80126, Naples, Italy
| | - Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 4, 80126, Naples, Italy
| |
Collapse
|
9
|
Nieto-Fabregat F, Marseglia A, Thépaut M, Kleman JP, Abbas M, Le Roy A, Ebel C, Maalej M, Simorre JP, Laguri C, Molinaro A, Silipo A, Fieschi F, Marchetti R. Molecular recognition of Escherichia coli R1-type core lipooligosaccharide by DC-SIGN. iScience 2024; 27:108792. [PMID: 38299112 PMCID: PMC10828809 DOI: 10.1016/j.isci.2024.108792] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Due to their ability to recognize carbohydrate structures, lectins emerged as potential receptors for bacterial lipopolysaccharides (LPS). Despite growing interest in investigating the association between host receptor lectins and exogenous glycan ligands, the molecular mechanisms underlying bacterial recognition by human lectins are still not fully understood. We contributed to fill this gap by unveiling the molecular basis of the interaction between the lipooligosaccharide of Escherichia coli and the dendritic cell-specific intracellular adhesion molecules (ICAM)-3 grabbing non-integrin (DC-SIGN). Specifically, a combination of different techniques, including fluorescence microscopy, surface plasmon resonance, NMR spectroscopy, and computational studies, demonstrated that DC-SIGN binds to the purified deacylated R1 lipooligosaccharide mainly through the recognition of its outer core pentasaccharide, which acts as a crosslinker between two different tetrameric units of DC-SIGN. Our results contribute to a better understanding of DC-SIGN-LPS interaction and may support the development of pharmacological and immunostimulatory strategies for bacterial infections, prevention, and therapy.
Collapse
Affiliation(s)
- Ferran Nieto-Fabregat
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Angela Marseglia
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Michel Thépaut
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Jean-Philippe Kleman
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Massilia Abbas
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Aline Le Roy
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Christine Ebel
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Meriem Maalej
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Jean-Pierre Simorre
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Cedric Laguri
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Antonio Molinaro
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Alba Silipo
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Franck Fieschi
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| | - Roberta Marchetti
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| |
Collapse
|
10
|
Elemam NM, Mekky RY, Rashid G, Braoudaki M, Youness RA. Pharmacogenomic and epigenomic approaches to untangle the enigma of IL-10 blockade in oncology. Expert Rev Mol Med 2024; 26:e1. [PMID: 38186186 PMCID: PMC10941350 DOI: 10.1017/erm.2023.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 01/09/2024]
Abstract
The host immune system status remains an unresolved mystery among several malignancies. An immune-compromised state or smart immune-surveillance tactics orchestrated by cancer cells are the primary cause of cancer invasion and metastasis. Taking a closer look at the tumour-immune microenvironment, a complex network and crosstalk between infiltrating immune cells and cancer cells mediated by cytokines, chemokines, exosomal mediators and shed ligands are present. Cytokines such as interleukins can influence all components of the tumour microenvironment (TME), consequently promoting or suppressing tumour invasion based on their secreting source. Interleukin-10 (IL-10) is an interlocked cytokine that has been associated with several types of malignancies and proved to have paradoxical effects. IL-10 has multiple functions on cellular and non-cellular components within the TME. In this review, the authors shed the light on the regulatory role of IL-10 in the TME of several malignant contexts. Moreover, detailed epigenomic and pharmacogenomic approaches for the regulation of IL-10 were presented and discussed.
Collapse
Affiliation(s)
- Noha M. Elemam
- Research Instiute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Radwa Y. Mekky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
| | - Gowhar Rashid
- Amity Medical School, Amity University, Gurugram (Manesar) 122413, Haryana, India
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
| | - Rana A. Youness
- Biology and Biochemistry Department, Faculty of Biotechnology, German International University, Cairo 11835, Egypt
| |
Collapse
|
11
|
McIntyre S, Warner J, Rush C, Vanderven HA. Antibodies as clinical tools for tuberculosis. Front Immunol 2023; 14:1278947. [PMID: 38162666 PMCID: PMC10755875 DOI: 10.3389/fimmu.2023.1278947] [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: 08/17/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Tuberculosis (TB) is a leading cause of morbidity and mortality worldwide. Global research efforts to improve TB control are hindered by insufficient understanding of the role that antibodies play in protective immunity and pathogenesis. This impacts knowledge of rational and optimal vaccine design, appropriate diagnostic biomarkers, and development of therapeutics. Traditional approaches for the prevention and diagnosis of TB may be less efficacious in high prevalence, remote, and resource-poor settings. An improved understanding of the immune response to the causative agent of TB, Mycobacterium tuberculosis (Mtb), will be crucial for developing better vaccines, therapeutics, and diagnostics. While memory CD4+ T cells and cells and cytokine interferon gamma (IFN-g) have been the main identified correlates of protection in TB, mounting evidence suggests that other types of immunity may also have important roles. TB serology has identified antibodies and functional characteristics that may help diagnose Mtb infection and distinguish between different TB disease states. To date, no serological tests meet the World Health Organization (WHO) requirements for TB diagnosis, but multiplex assays show promise for improving the sensitivity and specificity of TB serodiagnosis. Monoclonal antibody (mAb) therapies and serum passive infusion studies in murine models of TB have also demonstrated some protective outcomes. However, animal models that better reflect the human immune response to Mtb are necessary to fully assess the clinical utility of antibody-based TB prophylactics and therapeutics. Candidate TB vaccines are not designed to elicit an Mtb-specific antibody response, but evidence suggests BCG and novel TB vaccines may induce protective Mtb antibodies. The potential of the humoral immune response in TB monitoring and control is being investigated and these studies provide important insight into the functional role of antibody-mediated immunity against TB. In this review, we describe the current state of development of antibody-based clinical tools for TB, with a focus on diagnostic, therapeutic, and vaccine-based applications.
Collapse
Affiliation(s)
- Sophie McIntyre
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Jeffrey Warner
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Catherine Rush
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Hillary A. Vanderven
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
12
|
Kazemi S, Mirzaei R, Karampoor S, Hosseini-Fard SR, Ahmadyousefi Y, Soltanian AR, Keramat F, Saidijam M, Alikhani MY. Circular RNAs in tuberculosis: From mechanism of action to potential diagnostic biomarker. Microb Pathog 2023; 185:106459. [PMID: 37995882 DOI: 10.1016/j.micpath.2023.106459] [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: 05/25/2023] [Revised: 10/01/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tuberculosis), continues to be a major global health concern. Understanding the molecular intricacies of TB pathogenesis is crucial for developing effective diagnostic and therapeutic approaches. Circular RNAs (circRNAs), a class of single-stranded RNA molecules characterized by covalently closed loops, have recently emerged as potential diagnostic biomarkers in various diseases. CircRNAs have been demonstrated to modulate the host's immunological responses against TB, specifically by reducing monocyte apoptosis, augmenting autophagy, and facilitating macrophage polarization. This review comprehensively explores the roles and mechanisms of circRNAs in TB pathogenesis. We also discuss the growing body of evidence supporting their utility as promising diagnostic biomarkers for TB. By bridging the gap between fundamental circRNA biology and TB diagnostics, this review offers insights into the exciting potential of circRNAs in combatting this infectious disease.
Collapse
Affiliation(s)
- Sima Kazemi
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Reza Soltanian
- Department of Biostatistics and Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Iran
| | - Fariba Keramat
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
13
|
Abosalif KOA, Abdalla AE, Junaid K, Eltayeb LB, Ejaz H. The interleukin-10 family: Major regulators of the immune response against Plasmodium falciparum infections. Saudi J Biol Sci 2023; 30:103805. [PMID: 37727525 PMCID: PMC10506046 DOI: 10.1016/j.sjbs.2023.103805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
Malaria caused by the Plasmodium falciparum strain is more severe because of this protozoan's ability to disrupt the physiology of host cells during the blood stages of development by initiating the production of the interleukin-10 (IL-10) family of cytokines. P. falciparum feeds on hemoglobin and causes host cells to adhere to the walls of blood vessels by remodeling their composition. IL-10 is produced by CD4+ T cells that inhibits antigen-presenting cells' activity to prevent inflammation. This cytokine and its family members are crucial in promoting malarial infection by inhibiting the host's protective immune response, thus initiating Plasmodium parasitemia. IL-10 is also responsible for preventing severe pathology during Plasmodium infection and initiates several signaling pathways to alter the physiology of host cells during malarial infection. This review summarizes the critical aspects of P. falciparum infection, including its role in signaling pathways for cytokine exudation, its effect on microRNA, the human immune response in malaria, and the role played by the liver hormone hepcidin. Moreover, future aspects of vaccine development and therapeutic strategies to combat P. falciparum infections are also discussed in detail.
Collapse
Affiliation(s)
- Khalid Omer Abdalla Abosalif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abualgasim Elgaili Abdalla
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Kashaf Junaid
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Lienda Bashier Eltayeb
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin AbdulAziz University- Al-Kharj, 11942 Riyadh, Saudi Arabia
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| |
Collapse
|
14
|
van der Donk LEH, Bermejo-Jambrina M, van Hamme JL, Volkers MMW, van Nuenen AC, Kootstra NA, Geijtenbeek TBH. SARS-CoV-2 suppresses TLR4-induced immunity by dendritic cells via C-type lectin receptor DC-SIGN. PLoS Pathog 2023; 19:e1011735. [PMID: 37844099 PMCID: PMC10602378 DOI: 10.1371/journal.ppat.1011735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/26/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023] Open
Abstract
SARS-CoV-2 causes COVID-19, an infectious disease with symptoms ranging from a mild cold to severe pneumonia, inflammation, and even death. Although strong inflammatory responses are a major factor in causing morbidity and mortality, superinfections with bacteria during severe COVID-19 often cause pneumonia, bacteremia and sepsis. Aberrant immune responses might underlie increased sensitivity to bacteria during COVID-19 but the mechanisms remain unclear. Here we investigated whether SARS-CoV-2 directly suppresses immune responses to bacteria. We studied the functionality of human dendritic cells (DCs) towards a variety of bacterial triggers after exposure to SARS-CoV-2 Spike (S) protein and SARS-CoV-2 primary isolate (hCoV-19/Italy). Notably, pre-exposure of DCs to either SARS-CoV-2 S protein or a SARS-CoV-2 isolate led to reduced type I interferon (IFN) and cytokine responses in response to Toll-like receptor (TLR)4 agonist lipopolysaccharide (LPS), whereas other TLR agonists were not affected. SARS-CoV-2 S protein interacted with the C-type lectin receptor DC-SIGN and, notably, blocking DC-SIGN with antibodies restored type I IFN and cytokine responses to LPS. Moreover, blocking the kinase Raf-1 by a small molecule inhibitor restored immune responses to LPS. These results suggest that SARS-CoV-2 modulates DC function upon TLR4 triggering via DC-SIGN-induced Raf-1 pathway. These data imply that SARS-CoV-2 actively suppresses DC function via DC-SIGN, which might account for the higher mortality rates observed in patients with COVID-19 and bacterial superinfections.
Collapse
Affiliation(s)
- Lieve E. H. van der Donk
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Marta Bermejo-Jambrina
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - John L. van Hamme
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Mette M. W. Volkers
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Ad C. van Nuenen
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Neeltje A. Kootstra
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Teunis B. H. Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Zihad SNK, Sifat N, Islam MA, Monjur-Al-Hossain A, Sikdar KYK, Sarker MMR, Shilpi JA, Uddin SJ. Role of pattern recognition receptors in sensing Mycobacterium tuberculosis. Heliyon 2023; 9:e20636. [PMID: 37842564 PMCID: PMC10570006 DOI: 10.1016/j.heliyon.2023.e20636] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 09/06/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
Mycobacterium tuberculosis is one of the major invasive intracellular pathogens causing most deaths by a single infectious agent. The interaction between host immune cells and this pathogen is the focal point of the disease, Tuberculosis. Host immune cells not only mount the protective action against this pathogen but also serve as the primary niche for growth. Thus, recognition of this pathogen by host immune cells and following signaling cascades are key dictators of the disease state. Immune cells, mainly belonging to myeloid cell lineage, recognize a wide variety of Mycobacterium tuberculosis ligands ranging from carbohydrate and lipids to proteins to nucleic acids by different membrane-bound and soluble pattern recognition receptors. Simultaneous interaction between different host receptors and pathogen ligands leads to immune-inflammatory response as well as contributes to virulence. This review summarizes the contribution of pattern recognition receptors of host immune cells in recognizing Mycobacterium tuberculosis and subsequent initiation of signaling pathways to provide the molecular insight of the specific Mtb ligands interacting with specific PRR, key adaptor molecules of the downstream signaling pathways and the resultant effector functions which will aid in identifying novel drug targets, and developing novel drugs and adjuvants.
Collapse
Affiliation(s)
| | - Nazifa Sifat
- Department of Pharmacy, ASA University of Bangladesh, Dhaka, 1207, Bangladesh
| | | | | | | | - Md Moklesur Rahman Sarker
- Department of Pharmacy, State University of Bangladesh, Dhaka, 1205, Bangladesh
- Department of Pharmacy, Gono University, Nolam, Mirzanagar, Savar, Dhaka 1344, Bangladesh
| | - Jamil A. Shilpi
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
16
|
Pinho SS, Alves I, Gaifem J, Rabinovich GA. Immune regulatory networks coordinated by glycans and glycan-binding proteins in autoimmunity and infection. Cell Mol Immunol 2023; 20:1101-1113. [PMID: 37582971 PMCID: PMC10541879 DOI: 10.1038/s41423-023-01074-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023] Open
Abstract
The immune system is coordinated by an intricate network of stimulatory and inhibitory circuits that regulate host responses against endogenous and exogenous insults. Disruption of these safeguard and homeostatic mechanisms can lead to unpredictable inflammatory and autoimmune responses, whereas deficiency of immune stimulatory pathways may orchestrate immunosuppressive programs that contribute to perpetuate chronic infections, but also influence cancer development and progression. Glycans have emerged as essential components of homeostatic circuits, acting as fine-tuners of immunological responses and potential molecular targets for manipulation of immune tolerance and activation in a wide range of pathologic settings. Cell surface glycans, present in cells, tissues and the extracellular matrix, have been proposed to serve as "self-associated molecular patterns" that store structurally relevant biological data. The responsibility of deciphering this information relies on different families of glycan-binding proteins (including galectins, siglecs and C-type lectins) which, upon recognition of specific carbohydrate structures, can recalibrate the magnitude, nature and fate of immune responses. This process is tightly regulated by the diversity of glycan structures and the establishment of multivalent interactions on cell surface receptors and the extracellular matrix. Here we review the spatiotemporal regulation of selected glycan-modifying processes including mannosylation, complex N-glycan branching, core 2 O-glycan elongation, LacNAc extension, as well as terminal sialylation and fucosylation. Moreover, we illustrate examples that highlight the contribution of these processes to the control of immune responses and their integration with canonical tolerogenic pathways. Finally, we discuss the power of glycans and glycan-binding proteins as a source of immunomodulatory signals that could be leveraged for the treatment of autoimmune inflammation and chronic infection.
Collapse
Affiliation(s)
- Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal.
- Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal.
| | - Inês Alves
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Ciudad de Buenos Aires, Argentina.
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
17
|
Zhao L, Niu J, Feng D, Wang X, Zhang R. Immune functions of pattern recognition receptors in Lepidoptera. Front Immunol 2023; 14:1203061. [PMID: 37398667 PMCID: PMC10312389 DOI: 10.3389/fimmu.2023.1203061] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Pattern recognition receptors (PRRs), as the "sensors" in the immune response, play a prominent role in recognizing pathogen-associated molecular patterns (PAMPs) and initiating an effective defense response to pathogens in Lepidoptera. It is becoming increasingly clear that damage-associated molecular patterns (DAMPs) normally play a physiological role within cells; however, when exposed to extracellular, they may become "part-time" critical signals of the immune response. Based on research in recent years, we review herein typical PRRs of Lepidoptera, including peptidoglycan recognition protein (PGRP), gram-negative binding protein (GNBP), β-1,3-glucan recognition protein (βGRP), C-type lectin (CTL), and scavenger receptor (SR). We also outline the ways in which DAMPs participate in the immune response and the correlation between PRRs and immune escape. Taken together, these findings suggest that the role of PRRs in insect innate immunity may be much greater than expected and that it is possible to recognize a broader range of signaling molecules.
Collapse
Affiliation(s)
- Lin Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinlan Niu
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Disong Feng
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Xialu Wang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, China
| | - Rong Zhang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
18
|
Leusmann S, Ménová P, Shanin E, Titz A, Rademacher C. Glycomimetics for the inhibition and modulation of lectins. Chem Soc Rev 2023; 52:3663-3740. [PMID: 37232696 PMCID: PMC10243309 DOI: 10.1039/d2cs00954d] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 05/27/2023]
Abstract
Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.
Collapse
Affiliation(s)
- Steffen Leusmann
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Elena Shanin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
19
|
Viljoen A, Vercellone A, Chimen M, Gaibelet G, Mazères S, Nigou J, Dufrêne YF. Nanoscale clustering of mycobacterial ligands and DC-SIGN host receptors are key determinants for pathogen recognition. SCIENCE ADVANCES 2023; 9:eadf9498. [PMID: 37205764 PMCID: PMC10198640 DOI: 10.1126/sciadv.adf9498] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
The bacterial pathogen Mycobacterium tuberculosis binds to the C-type lectin DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) on dendritic cells to evade the immune system. While DC-SIGN glycoconjugate ligands are ubiquitous among mycobacterial species, the receptor selectively binds pathogenic species from the M. tuberculosis complex (MTBC). Here, we unravel the molecular mechanism behind this intriguing selective recognition by means of a multidisciplinary approach combining single-molecule atomic force microscopy with Förster resonance energy transfer and bioassays. Molecular recognition imaging of mycobacteria demonstrates that the distribution of DC-SIGN ligands markedly differs between Mycobacterium bovis Bacille Calmette-Guérin (BCG) (model MTBC species) and Mycobacterium smegmatis (non-MTBC species), the ligands being concentrated into dense nanodomains on M. bovis BCG. Upon bacteria-host cell adhesion, ligand nanodomains induce the recruitment and clustering of DC-SIGN. Our study highlights the key role of clustering of both ligands on MTBC species and DC-SIGN host receptors in pathogen recognition, a mechanism that might be widespread in host-pathogen interactions.
Collapse
Affiliation(s)
- Albertus Viljoen
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07., B-1348 Louvain-la-Neuve, Belgium
| | - Alain Vercellone
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Myriam Chimen
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Gérald Gaibelet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Yves F. Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07., B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
20
|
Papin L, Lehmann M, Lagisquet J, Maarifi G, Robert-Hebmann V, Mariller C, Guerardel Y, Espert L, Haucke V, Blanchet FP. The Autophagy Nucleation Factor ATG9 Forms Nanoclusters with the HIV-1 Receptor DC-SIGN and Regulates Early Antiviral Autophagy in Human Dendritic Cells. Int J Mol Sci 2023; 24:ijms24109008. [PMID: 37240354 DOI: 10.3390/ijms24109008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Dendritic cells (DC) are critical cellular mediators of host immunity, notably by expressing a broad panel of pattern recognition receptors. One of those receptors, the C-type lectin receptor DC-SIGN, was previously reported as a regulator of endo/lysosomal targeting through functional connections with the autophagy pathway. Here, we confirmed that DC-SIGN internalization intersects with LC3+ autophagy structures in primary human monocyte-derived dendritic cells (MoDC). DC-SIGN engagement promoted autophagy flux which coincided with the recruitment of ATG-related factors. As such, the autophagy initiation factor ATG9 was found to be associated with DC-SIGN very early upon receptor engagement and required for an optimal DC-SIGN-mediated autophagy flux. The autophagy flux activation upon DC-SIGN engagement was recapitulated using engineered DC-SIGN-expressing epithelial cells in which ATG9 association with the receptor was also confirmed. Finally, Stimulated emission depletion (STED) microscopy performed in primary human MoDC revealed DC-SIGN-dependent submembrane nanoclusters formed with ATG9, which was required to degrade incoming viruses and further limit DC-mediated transmission of HIV-1 infection to CD4+ T lymphocytes. Our study unveils a physical association between the Pattern Recognition Receptor DC-SIGN and essential components of the autophagy pathway contributing to early endocytic events and the host's antiviral immune response.
Collapse
Affiliation(s)
- Laure Papin
- Institut de Recherche en Infectiologie de Montpellier-IRIM-CNRS UMR9004, University of Montpellier, 34090 Montpellier, France
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Justine Lagisquet
- Institut de Recherche en Infectiologie de Montpellier-IRIM-CNRS UMR9004, University of Montpellier, 34090 Montpellier, France
| | - Ghizlane Maarifi
- Institut de Recherche en Infectiologie de Montpellier-IRIM-CNRS UMR9004, University of Montpellier, 34090 Montpellier, France
| | - Véronique Robert-Hebmann
- Institut de Recherche en Infectiologie de Montpellier-IRIM-CNRS UMR9004, University of Montpellier, 34090 Montpellier, France
| | - Christophe Mariller
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Yann Guerardel
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1112, Japan
| | - Lucile Espert
- Institut de Recherche en Infectiologie de Montpellier-IRIM-CNRS UMR9004, University of Montpellier, 34090 Montpellier, France
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Fabien P Blanchet
- Institut de Recherche en Infectiologie de Montpellier-IRIM-CNRS UMR9004, University of Montpellier, 34090 Montpellier, France
| |
Collapse
|
21
|
Gong Y, Wang J, Li F, Zhu B. Polysaccharides and glycolipids of Mycobacterium tuberculosis and their induced immune responses. Scand J Immunol 2023; 97:e13261. [PMID: 39008002 DOI: 10.1111/sji.13261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 07/16/2024]
Abstract
Tuberculosis (TB) is a chronic infectious disease mainly caused by Mycobacterium tuberculosis (M. tuberculosis). The structures of polysaccharides and glycolipids at M. tuberculosis cell wall vary among different strains, which affect the physiology and pathogenesis of mycobacteria by activating or inhibiting innate and acquired immunity. Among them, some components such as lipomannan (LM) and lipoarabinomannan (LAM) activate innate immunity by recognizing some kinds of pattern recognition receptors (PRRs) like Toll-like receptors, while other components like mannose-capped lipoarabinomannan (ManLAM) could prevent innate immune responses by inhibiting the secretion of pro-inflammatory cytokines and maturation of phagosomes. In addition, many glycolipids can activate natural killer T (NKT) cells and CD1-restricted T cells to produce interferon-γ (IFN-γ). Furthermore, humoral immunity against cell wall components, such as antibodies against LAM, plays a role in immunity against M. tuberculosis infection. Cell wall polysaccharides and glycolipids of M. tuberculosis have potential applications as antigens and adjuvants for novel TB subunit vaccines.
Collapse
Affiliation(s)
- Yang Gong
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Juan Wang
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fei Li
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
22
|
Cummings MJ, Bakamutumaho B, Jain K, Price A, Owor N, Kayiwa J, Namulondo J, Byaruhanga T, Muwanga M, Nsereko C, Nayiga I, Kyebambe S, Che X, Sameroff S, Tokarz R, Wong W, Postler TS, Larsen MH, Lipkin WI, Lutwama JJ, O’Donnell MR. Brief Report: Detection of Urine Lipoarabinomannan Is Associated With Proinflammatory Innate Immune Activation, Impaired Host Defense, and Organ Dysfunction in Adults With Severe HIV-Associated Tuberculosis in Uganda. J Acquir Immune Defic Syndr 2023; 93:79-85. [PMID: 36701194 PMCID: PMC10079575 DOI: 10.1097/qai.0000000000003159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND The immunopathology of disseminated HIV-associated tuberculosis (HIV/TB), a leading cause of critical illness and death among persons living with HIV in sub-Saharan Africa, is incompletely understood. Reflective of hematogenously disseminated TB, detection of lipoarabinomannan (LAM) in urine is associated with greater bacillary burden and poor outcomes in adults with HIV/TB. METHODS We determined the relationship between detection of urine TB-LAM, organ dysfunction, and host immune responses in a prospective cohort of adults hospitalized with severe HIV/TB in Uganda. Generalized additive models were used to analyze the association between urine TB-LAM grade and concentrations of 14 soluble immune mediators. Whole-blood RNA-sequencing data were used to compare transcriptional profiles between patients with high- vs. low-grade TB-LAM results. RESULTS Among 157 hospitalized persons living with HIV, 40 (25.5%) had positive urine TB-LAM testing. Higher TB-LAM grade was associated with more severe physiologic derangement, organ dysfunction, and shock. Adjusted generalized additive models showed that higher TB-LAM grade was significantly associated with higher concentrations of mediators reflecting proinflammatory innate and T-cell activation and chemotaxis (IL-8, MIF, MIP-1β/CCL4, and sIL-2Ra/sCD25). Transcriptionally, patients with higher TB-LAM grades demonstrated multifaceted impairment of antibacterial defense including reduced expression of genes encoding cytotoxic and autophagy-related proteins and impaired cross-talk between innate and cell-mediated immune effectors. CONCLUSIONS Our findings add to emerging data suggesting pathobiological relationships between LAM, TB dissemination, innate cell activation, and evasion of host immunity in severe HIV/TB. Further translational studies are needed to elucidate the role for immunomodulatory therapies, in addition to optimized anti-TB treatment, in this often critically ill population.
Collapse
Affiliation(s)
- Matthew J. Cummings
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Barnabas Bakamutumaho
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
- Immunizable Diseases Unit, Uganda Virus Research Institute, Entebbe, Uganda
| | - Komal Jain
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Adam Price
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Nicholas Owor
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - John Kayiwa
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Joyce Namulondo
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Timothy Byaruhanga
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Moses Muwanga
- Entebbe General Referral Hospital, Ministry of Health, Entebbe, Uganda
| | | | - Irene Nayiga
- Entebbe General Referral Hospital, Ministry of Health, Entebbe, Uganda
| | - Stephen Kyebambe
- Entebbe General Referral Hospital, Ministry of Health, Entebbe, Uganda
| | - Xiaoyu Che
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Stephen Sameroff
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Rafal Tokarz
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Wai Wong
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Thomas S. Postler
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Michelle H. Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - W. Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Julius J. Lutwama
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Max R. O’Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
23
|
Miller MH, Swaby LG, Vailoces VS, LaFratta M, Zhang Y, Zhu X, Hitchcock DJ, Jewett TJ, Zhang B, Tigno-Aranjuez JT. LMAN1 is a receptor for house dust mite allergens. Cell Rep 2023; 42:112208. [PMID: 36870056 PMCID: PMC10105285 DOI: 10.1016/j.celrep.2023.112208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/01/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Development of therapies with the potential to change the allergic asthmatic disease course will require the discovery of targets that play a central role during the initiation of an allergic response, such as those involved in the process of allergen recognition. We use a receptor glycocapture technique to screen for house dust mite (HDM) receptors and identify LMAN1 as a candidate. We verify the ability of LMAN1 to directly bind HDM allergens and demonstrate that LMAN1 is expressed on the surface of dendritic cells (DCs) and airway epithelial cells (AECs) in vivo. Overexpression of LMAN1 downregulates NF-κB signaling in response to inflammatory cytokines or HDM. HDM promotes binding of LMAN1 to the FcRγ and recruitment of SHP1. Last, peripheral DCs of asthmatic individuals show a significant reduction in the expression of LMAN1 compared with healthy controls. These findings have potential implications for the development of therapeutic interventions for atopic disease.
Collapse
Affiliation(s)
- Madelyn H Miller
- Biotechnology and Immunology Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Lindsay G Swaby
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Vanessa S Vailoces
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Maggie LaFratta
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Yuan Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Xiang Zhu
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Dorilyn J Hitchcock
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Travis J Jewett
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Justine T Tigno-Aranjuez
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| |
Collapse
|
24
|
Santiago KB, Conti BJ, Cardoso EDO, Conte FL, Tasca KI, Romagnoli GG, Golim MDA, Cruz MT, Sforcin JM. Propolis anti-inflammatory effects on MAGE-1 and retinoic acid-treated dendritic cells and on Th1 and T regulatory cells. J Venom Anim Toxins Incl Trop Dis 2023; 29:e20220044. [PMID: 36721426 PMCID: PMC9851646 DOI: 10.1590/1678-9199-jvatitd-2022-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/13/2022] [Indexed: 01/15/2023] Open
Abstract
Background Propolis exhibits huge potential in the pharmaceutical industry. In the present study, its effects were investigated on dendritic cells (DCs) stimulated with a tumor antigen (MAGE-1) and retinoic acid (RA) and on T lymphocytes to observe a possible differential activation of T lymphocytes, driving preferentially to Th1 or Treg cells. Methods Cell viability, lymphocyte proliferation, gene expression (T-bet and FoxP3), and cytokine production by DCs (TNF-α, IL-10, IL-6 and IL-1β) and lymphocytes (IFN-γ and TGF-β) were analyzed. Results MAGE-1 and RA alone or in combination with propolis inhibited TNF-α production and induced a higher lymphoproliferation compared to control, while MAGE-1 + propolis induced IL-6 production. Propolis in combination with RA induced FoxP3 expression. MAGE-1 induced IFN-γ production while propolis inhibited it, returning to basal levels. RA inhibited TGF-β production, what was counteracted by propolis. Conclusion Propolis affected immunological parameters inhibiting pro-inflammatory cytokines and favoring the regulatory profile, opening perspectives for the control of inflammatory conditions.
Collapse
Affiliation(s)
| | - Bruno José Conti
- Institute of Biosciences, São Paulo State University (UNESP),
Botucatu, SP, Brazil
| | | | - Fernanda Lopes Conte
- Institute of Biosciences, São Paulo State University (UNESP),
Botucatu, SP, Brazil
| | - Karen Ingrid Tasca
- Institute of Biosciences, São Paulo State University (UNESP),
Botucatu, SP, Brazil
| | | | | | - Maria Tereza Cruz
- Faculty of Pharmacy, Center for Neurosciences and Cellular Biology,
University of Coimbra, Coimbra, Portugal
| | - José Maurício Sforcin
- Institute of Biosciences, São Paulo State University (UNESP),
Botucatu, SP, Brazil.,Correspondence:
| |
Collapse
|
25
|
Schick J, Altunay M, Lacorcia M, Marschner N, Westermann S, Schluckebier J, Schubart C, Bodendorfer B, Christensen D, Alexander C, Wirtz S, Voehringer D, da Costa CP, Lang R. IL-4 and helminth infection downregulate MINCLE-dependent macrophage response to mycobacteria and Th17 adjuvanticity. eLife 2023; 12:72923. [PMID: 36753434 PMCID: PMC9908076 DOI: 10.7554/elife.72923] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
The myeloid C-type lectin receptor (CLR) MINCLE senses the mycobacterial cell wall component trehalose-6,6'-dimycolate (TDM). Recently, we found that IL-4 downregulates MINCLE expression in macrophages. IL-4 is a hallmark cytokine in helminth infections, which appear to increase the risk for mycobacterial infection and active tuberculosis. Here, we investigated functional consequences of IL-4 and helminth infection on MINCLE-driven macrophage activation and Th1/Th17 adjuvanticity. IL-4 inhibited MINCLE and cytokine induction after macrophage infection with Mycobacterium bovis bacille Calmette-Guerin (BCG). Infection of mice with BCG upregulated MINCLE on myeloid cells, which was inhibited by IL-4 plasmid injection and by infection with the nematode Nippostrongylus brasiliensis in monocytes. To determine the impact of helminth infection on MINCLE-dependent immune responses, we vaccinated mice with a recombinant protein together with the MINCLE ligand trehalose-6,6-dibehenate (TDB) as adjuvant. Concurrent infection with N. brasiliensis or with Schistosoma mansoni promoted T cell-derived IL-4 production and suppressed Th1/Th17 differentiation in the spleen. In contrast, helminth infection did not reduce Th1/Th17 induction by TDB in draining peripheral lymph nodes, where IL-4 levels were unaltered. Upon use of the TLR4-dependent adjuvant G3D6A, N. brasiliensis infection impaired selectively the induction of splenic antigen-specific Th1 but not of Th17 cells. Inhibition of MINCLE-dependent Th1/Th17 responses in mice infected with N. brasiliensis was dependent on IL-4/IL-13. Thus, helminth infection attenuated the Th17 response to MINCLE-dependent immunization in an organ- and adjuvant-specific manner via the Th2 cytokines IL-4/IL-13. Taken together, our results demonstrate downregulation of MINCLE expression on monocytes and macrophages by IL-4 as a possible mechanism of thwarted Th17 vaccination responses by underlying helminth infection.
Collapse
Affiliation(s)
- Judith Schick
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Meltem Altunay
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Matthew Lacorcia
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Center for Global Health, Technische Universität MünchenMunichGermany,Center for Global Health, Technical University MunichMunichGermany
| | - Nathalie Marschner
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Stefanie Westermann
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Julia Schluckebier
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Center for Global Health, Technische Universität MünchenMunichGermany,Center for Global Health, Technical University MunichMunichGermany
| | - Christoph Schubart
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Barbara Bodendorfer
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Dennis Christensen
- Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum InstitutCopenhagenDenmark
| | - Christian Alexander
- Cellular Microbiology, Forschungszentrum Borstel, Leibniz Lung Center BorstelBorstelGermany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - David Voehringer
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| | - Clarissa Prazeres da Costa
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Center for Global Health, Technische Universität MünchenMunichGermany,Center for Global Health, Technical University MunichMunichGermany
| | - Roland Lang
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-NürnbergErlangenGermany
| |
Collapse
|
26
|
Conti BJ, Santiago KB, Cardoso EO, Conte FL, Golim MA, Cruz MT, Sforcin JM. Effect of propolis on Th2 and Th17 cells: interplay with EtxB- and LPS-treated dendritic cells. Braz J Med Biol Res 2023; 56:e12659. [PMID: 37075347 PMCID: PMC10125804 DOI: 10.1590/1414-431x2023e12659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/07/2023] [Indexed: 04/21/2023] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that drive the differentiation of T CD4+ cells into different profiles according to the nature of the antigen or immunomodulator. Propolis is a resinous product made by bees that has numerous pharmacological properties, including an immunomodulatory action. To assess whether propolis can modulate the activation of CD4+ T cells by stimulating DCs with heat-labile enterotoxin B subunit (EtxB) or lipopolysaccharide (LPS), we aimed to elucidate the mechanisms affected by propolis in the differential activation of T lymphocytes. Cell viability, lymphocyte proliferation, gene expression (GATA-3 and RORc), and cytokine production (interleukin (IL)-4 and IL-17A) were analyzed. Propolis, EtxB, and LPS induced a higher lymphoproliferation compared with the control. Propolis induced GATA-3 expression and, in combination with EtxB, maintained the baseline levels. Propolis alone or in combination with LPS inhibited RORc expression. EtxB alone and in combination with propolis increased IL-4 production. Propolis in combination with LPS prevented LPS-induced IL-17A production. These results opened perspectives for the study of biological events that may be favored by propolis by promoting Th2 activation or helping in the treatment of inflammatory conditions mediated by Th17 cells.
Collapse
Affiliation(s)
- B J Conti
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - K B Santiago
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - E O Cardoso
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - F L Conte
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - M A Golim
- Hemocentro de Botucatu, Faculdade de Medicina, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - M T Cruz
- Faculty of Pharmacy, Center for Neurosciences and Cellular Biology, University of Coimbra, Coimbra, Portugal
| | - J M Sforcin
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| |
Collapse
|
27
|
Correia-Neves M, Nigou J, Mousavian Z, Sundling C, Källenius G. Immunological hyporesponsiveness in tuberculosis: The role of mycobacterial glycolipids. Front Immunol 2022; 13:1035122. [PMID: 36544778 PMCID: PMC9761185 DOI: 10.3389/fimmu.2022.1035122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 12/09/2022] Open
Abstract
Glycolipids constitute a major part of the cell envelope of Mycobacterium tuberculosis (Mtb). They are potent immunomodulatory molecules recognized by several immune receptors like pattern recognition receptors such as TLR2, DC-SIGN and Dectin-2 on antigen-presenting cells and by T cell receptors on T lymphocytes. The Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic relatives, phosphatidylinositol mannosides (PIMs) and lipomannan (LM), as well as other Mtb glycolipids, such as phenolic glycolipids and sulfoglycolipids have the ability to modulate the immune response, stimulating or inhibiting a pro-inflammatory response. We explore here the downmodulating effect of Mtb glycolipids. A great proportion of the studies used in vitro approaches although in vivo infection with Mtb might also lead to a dampening of myeloid cell and T cell responses to Mtb glycolipids. This dampened response has been explored ex vivo with immune cells from peripheral blood from Mtb-infected individuals and in mouse models of infection. In addition to the dampening of the immune response caused by Mtb glycolipids, we discuss the hyporesponse to Mtb glycolipids caused by prolonged Mtb infection and/or exposure to Mtb antigens. Hyporesponse to LAM has been observed in myeloid cells from individuals with active and latent tuberculosis (TB). For some myeloid subsets, this effect is stronger in latent versus active TB. Since the immune response in individuals with latent TB represents a more protective profile compared to the one in patients with active TB, this suggests that downmodulation of myeloid cell functions by Mtb glycolipids may be beneficial for the host and protect against active TB disease. The mechanisms of this downmodulation, including tolerance through epigenetic modifications, are only partly explored.
Collapse
Affiliation(s)
- Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal,Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B's), Portuguese (PT) Government Associate Laboratory, Braga, Portugal,Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France
| | - Zaynab Mousavian
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,School of Mathematics, Statistics, and Computer Science, College of Science, University of Tehran, Tehran, Iran,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,*Correspondence: Gunilla Källenius,
| |
Collapse
|
28
|
Krivonogova AS, Bruter AV, Makutina VA, Okulova YD, Ilchuk LA, Kubekina MV, Khamatova AY, Egorova TV, Mymrin VS, Silaeva YY, Deykin AV, Filatov MA, Isaeva AG. AAV infection of bovine embryos: Novel, simple and effective tool for genome editing. Theriogenology 2022; 193:77-86. [PMID: 36156427 DOI: 10.1016/j.theriogenology.2022.09.007] [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: 04/26/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/28/2022]
Abstract
Adeno-associated viruses (AAV) are widely used in the field of genetically modified organism production. In this work, transduction of bovine embryos by AAV was selected as a potential approach to perform genetic modifications: we have used recombinant AAV to produce GFP-positive bovine embryos. Five different AAV serotypes were used to evaluate their ability to deliver genetic material into the bovine embryos. AAV9 serotype demonstrated minimal effectiveness (38,10%) as the genetic material transfer tool. Four other serotypes of AAVs (AAV1, AAV2, AAV6 and AAV-DJ) showed very close transduction efficiency (52,94-58,33%). CD209 is a C-type lectin receptor which is presented on the surface of macrophages and dendritic cells. CD209 recognizes a broad range of pathogens in a rather nonspecific manner. Production of CD209 knock-out is relevant for better understanding of infection mechanisms. Potentially, production of such knock-out may enable animals to become resistant to various infections. We have analyzed DNA samples from 22 blastocysts obtained after in vitro culture of zygotes subjected to recombinant AAV action. We have detected that 3 of 22 analyzed blastocysts contained mosaic CD209 frameshifts. Therefore, we have demonstrated proof of principle that application of AAV as a genome editing tool is an effective method for obtaining genetically modified cattle embryos.
Collapse
Affiliation(s)
- Anna S Krivonogova
- Ural Federal Agrarian Research Center of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Alexandra V Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Valeria A Makutina
- Ural Federal Agrarian Research Center of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Yuliya D Okulova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Leonid A Ilchuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Marina V Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Alexandra Yu Khamatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Tatiana V Egorova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia; Marlin Biotech LLC, Sochi, 354340, Russia
| | - Vladimir S Mymrin
- Ural Federal Agrarian Research Center of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Yuliya Yu Silaeva
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Alexey V Deykin
- Ural Federal Agrarian Research Center of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Maxim A Filatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Albina G Isaeva
- Ural Federal Agrarian Research Center of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| |
Collapse
|
29
|
Olejnik B, Ferens-Sieczkowska M. Seminal Plasma Glycoproteins as Potential Ligands of Lectins Engaged in Immunity Regulation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10489. [PMID: 36078205 PMCID: PMC9518496 DOI: 10.3390/ijerph191710489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Environmental pollution, chronic stress, and unhealthy lifestyle are factors that negatively affect reproductive potential. Currently, 15-20% of couples in industrialized countries face the problem of infertility. This growing health and social problem prompts researchers to explore the regulatory mechanisms that may be important for successful fertilization. In recent years, more attention has been paid to male infertility factors, including the impact of seminal plasma components on regulation of the female immune response to allogenic sperm, embryo and fetal antigens. Directing this response to the tolerogenic pathway is crucial to achieve a healthy pregnancy. According to the fetoembryonic defense hypothesis, the regulatory mechanism may be associated with the interaction of lectins and immunomodulatory glycoepitopes. Such interactions may involve lectins of dendritic cells and macrophages, recruited to the cervical region immediately after intercourse. Carbohydrate binding receptors include C type lectins, such as DC-SIGN and MGL, as well as galectins and siglecs among others. In this article we discuss the expression of the possible lectin ligands, highly fucosylated and high mannose structures, which may be recognized by DC-SIGN, glycans of varying degrees of sialylation, which may differ in their interaction with siglecs, as well as T and Tn antigens in O-glycans.
Collapse
|
30
|
Abstract
Macrophage surface receptors are critical for pathogen defense, as they are the gatekeepers for pathogen entry and sensing, which trigger robust immune responses. TREM2 (triggering receptor expressed on myeloid cells 2) is a transmembrane surface receptor that mediates anti-inflammatory immune signaling. A recent study showed that TREM2 is a receptor for mycolic acids in the mycobacterial cell wall and inhibits macrophage activation. However, the underlying functional mechanism of how TREM2 regulates the macrophage antimycobacterial response remains unclear. Here, we show that Mycobacterium tuberculosis, the causative agent for tuberculosis, specifically binds to human TREM2 to disable the macrophage antibacterial response. Live but not killed mycobacteria specifically trigger the upregulation of TREM2 during macrophage infection through a mechanism dependent on STING (the stimulator of interferon genes). TREM2 facilitated uptake of M. tuberculosis into macrophages and is responsible for blocking the production of tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), and reactive oxygen species (ROS), while enhancing the production of interferon-β (IFN-β) and IL-10. TREM2-mediated blockade of ROS production promoted the survival of M. tuberculosis within infected macrophages. Consistent with this, genetic deletion or antibody-mediated neutralization of TREM2 reduced the intracellular survival of M. tuberculosis through enhanced production of ROS. Importantly, inhibition of type I IFN signaling in TREM2-overexpressing macrophages restored the ability of these cells to produce inflammatory cytokines and ROS, resulting in normal levels of intracellular bacteria killing. Collectively, our study identifies TREM2 as an attractive host receptor for host-directed antimycobacterial therapeutics. IMPORTANCE Mycobacterium tuberculosis is one of the most ancient bacterial pathogens and remains the leading cause of death from a single bacterial agent. The success of M. tuberculosis relies greatly on its ability to parasitize and disable its host macrophages. Previous studies have found that M. tuberculosis uses its unique cell wall lipids to manipulate the immune response by binding to specific surface receptors on macrophages. Our study reveals that M. tuberculosis binds to TREM2, an immunomodulatory receptor expressed on macrophages, to facilitate a "silent" mode of entry. Increased levels of TREM2 triggered by intracellular sensing of M. tuberculosis promoted the intracellular survival of M. tuberculosis through type I IFN-driven inhibition of reactive oxygen species (ROS) and proinflammatory cytokine production. Importantly, deletion of TREM2 reversed the effects of "silent" entry and resulted in increased production of inflammatory cytokines, generation of ROS, and cell death. As such, antibody-mediated or pharmacological targeting of TREM2 could be a promising strategy for novel treatments against M. tuberculosis infection.
Collapse
|
31
|
Kim H, Shin SJ. Pathological and protective roles of dendritic cells in Mycobacterium tuberculosis infection: Interaction between host immune responses and pathogen evasion. Front Cell Infect Microbiol 2022; 12:891878. [PMID: 35967869 PMCID: PMC9366614 DOI: 10.3389/fcimb.2022.891878] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) are principal defense components that play multifactorial roles in translating innate immune responses to adaptive immunity in Mycobacterium tuberculosis (Mtb) infections. The heterogeneous nature of DC subsets follows their altered functions by interacting with other immune cells, Mtb, and its products, enhancing host defense mechanisms or facilitating pathogen evasion. Thus, a better understanding of the immune responses initiated, promoted, and amplified or inhibited by DCs in Mtb infection is an essential step in developing anti-tuberculosis (TB) control measures, such as host-directed adjunctive therapy and anti-TB vaccines. This review summarizes the recent advances in salient DC subsets, including their phenotypic classification, cytokine profiles, functional alterations according to disease stages and environments, and consequent TB outcomes. A comprehensive overview of the role of DCs from various perspectives enables a deeper understanding of TB pathogenesis and could be useful in developing DC-based vaccines and immunotherapies.
Collapse
|
32
|
Arnold JN, Mitchell DA. Tinker, tailor, soldier, cell: the role of C-type lectins in the defense and promotion of disease. Protein Cell 2022; 14:4-16. [PMID: 36726757 PMCID: PMC9871964 DOI: 10.1093/procel/pwac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
C-type lectins (CTLs) represent a large family of soluble and membrane-bound proteins which bind calcium dependently via carbohydrate recognition domains (CRDs) to glycan residues presented on the surface of a variety of pathogens. The deconvolution of a cell's glycan code by CTLs underpins several important physiological processes in mammals such as pathogen neutralization and opsonization, leukocyte trafficking, and the inflammatory response. However, as our knowledge of CTLs has developed it has become apparent that the role of this innate immune family of proteins can be double-edged, where some pathogens have developed approaches to subvert and exploit CTL interactions to promote infection and sustain the pathological state. Equally, CTL interactions with host glycoproteins can contribute to inflammatory diseases such as arthritis and cancer whereby, in certain contexts, they exacerbate inflammation and drive malignant progression. This review discusses the 'dual agent' roles of some of the major mammalian CTLs in both resolving and promoting infection, inflammation and inflammatory disease and highlights opportunities and emerging approaches for their therapeutic modulation.
Collapse
|
33
|
Cagliero J, Vernel-Pauillac F, Murray G, Adler B, Matsui M, Werts C. Pathogenic Leptospires Limit Dendritic Cell Activation Through Avoidance of TLR4 and TRIF Signaling. Front Immunol 2022; 13:911778. [PMID: 35812397 PMCID: PMC9258186 DOI: 10.3389/fimmu.2022.911778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Leptospira interrogans is a bacterial species responsible for leptospirosis, a neglected worldwide zoonosis. Mice and rats are resistant and can become asymptomatic carriers, whereas humans and some other mammals may develop severe forms of leptospirosis. Uncommon among spirochetes, leptospires contain lipopolysaccharide (LPS) in their outer membrane. LPS is highly immunogenic and forms the basis for a large number of serovars. Vaccination with inactivated leptospires elicits a protective immunity, restricted to serovars with related LPS. This protection that lasts in mice, is not long lasting in humans and requires annual boosts. Leptospires are stealth pathogens that evade the complement system and some pattern recognition receptors from the Toll-like (TLR) and Nod-Like families, therefore limiting antibacterial defense. In macrophages, leptospires totally escape recognition by human TLR4, and escape the TRIF arm of the mouse TLR4 pathway. However, very little is known about the recognition and processing of leptospires by dendritic cells (DCs), although they are crucial cells linking innate and adaptive immunity. Here we tested the activation of primary DCs derived from human monocytes (MO-DCs) and mouse bone marrow (BM-DCs) 24h after stimulation with saprophytic or different pathogenic virulent or avirulent L. interrogans. We measured by flow cytometry the expression of DC-SIGN, a lectin involved in T-cell activation, co-stimulation molecules and MHC-II markers, and pro- and anti-inflammatory cytokines by ELISA. We found that exposure to leptospires, live or heat-killed, activated dendritic cells. However, pathogenic L. interrogans, especially from the Icterohaemorraghiae Verdun strain, triggered less marker upregulation and less cytokine production than the saprophytic Leptospira biflexa. In addition, we showed a better activation with avirulent leptospires, when compared to the virulent parental strains in murine BM-DCs. We did not observe this difference in human MO-DCs, suggesting a role for TLR4 in DC stimulation. Accordingly, using BM-DCs from transgenic deficient mice, we showed that virulent Icterohaemorraghiae and Manilae serovars dampened DC activation, at least partly, through the TLR4 and TRIF pathways. This work shows a novel bacterial immune evasion mechanism to limit DC activation and further illustrates the role of the leptospiral LPS as a virulence factor.
Collapse
Affiliation(s)
- Julie Cagliero
- Institut Pasteur de Nouvelle-Calédonie, member of the Pasteur Network, Immunity and Inflammation Group (GIMIN), Noumea, New Caledonia
- Institut Pasteur, Université de Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi bactérienne, F-75015 Paris, France
- Institut Pasteur de Nouvelle-Calédonie, member of the Pasteur Network, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Université de Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi bactérienne, F-75015 Paris, France
| | - Gerald Murray
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Ben Adler
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Mariko Matsui
- Institut Pasteur de Nouvelle-Calédonie, member of the Pasteur Network, Immunity and Inflammation Group (GIMIN), Noumea, New Caledonia
| | - Catherine Werts
- Institut Pasteur, Université de Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi bactérienne, F-75015 Paris, France
- *Correspondence: Catherine Werts,
| |
Collapse
|
34
|
Germano GV, Braga AF, Camargo RMD, Ballalai PB, Bezerra OC, Manta FSDN, Belone ADFF, Soares CT, Das PK, Moraes MO, Latini ACP, Brito de Souza VN. Association of CD209 (DC-SIGN) rs735240 SNV with paucibacillary leprosy in the Brazilian population and its functional effects. Mem Inst Oswaldo Cruz 2022; 117:e220014. [PMID: 35703715 PMCID: PMC9190517 DOI: 10.1590/0074-02760220014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Leprosy, caused by Mycobacterium leprae, is a public health problem in Brazil that affects peripheral nerves, resulting in physical disabilities. During host-pathogen interactions, the immune response determines leprosy outcomes from a localised (paucibacillary) form to a disseminated (multibacillary) form. The recognition of M. leprae involves the DC-SIGN receptor, which is present on the dendritic cells (DCs) and participates in immune activation. OBJECTIVES To evaluate the association of polymorphisms in the promoter region of the gene encoding DC-SIGN (CD209) and the clinical form of leprosy, and to investigate its functional effects. METHODS The study population included 406 leprosy patients from an endemic area in Brazil [310 multibacillary (MB); 96 paucibacillary (PB)]. A functional evaluation based on the effects of the single nucleotide variant (SNV) associated with PB leprosy on the specific immune response was also performed. RESULTS The GA genotype and the presence of the A allele of rs735240 (-939G>A) were associated with PB leprosy [OR: 2.09 (1.18-3.69) and 1.84 (1.07-3.14), respectively]. Carriers of the A allele showed reduced expression of CD209 and TGF-β1 in leprosy lesions in comparison with individuals with GG genotype, in addition to a higher response to the Mitsuda test. CONCLUSION These data suggest that rs735240 influences the immune response against M. leprae and clinical presentation of leprosy.
Collapse
Affiliation(s)
- Giovanna Valle Germano
- Faculdade de Medicina de Botucatu, Programa de Pós-Graduação em Doenças Tropicais, Botucatu, SP, Brasil.,Secretaria de Estado da Saúde de São Paulo, Instituto Lauro de Souza Lima, Bauru, SP, Brasil
| | - André Flores Braga
- Faculdade de Medicina de Botucatu, Programa de Pós-Graduação em Doenças Tropicais, Botucatu, SP, Brasil.,Secretaria de Estado da Saúde de São Paulo, Instituto Lauro de Souza Lima, Bauru, SP, Brasil
| | - Rodrigo Mendes de Camargo
- Faculdade de Medicina de Botucatu, Programa de Pós-Graduação em Doenças Tropicais, Botucatu, SP, Brasil.,Secretaria de Estado da Saúde de São Paulo, Instituto Lauro de Souza Lima, Bauru, SP, Brasil
| | | | - Ohanna Cavalcanti Bezerra
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Hanseníase, Rio de Janeiro, RJ, Brasil
| | | | | | | | - Pranab Kumar Das
- University of Birmingham, College of Medical and Dental Sciences, Division of Infection and Immunity, Department of Clinical Immunology, Edgbaston, UK
| | - Milton Ozório Moraes
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Hanseníase, Rio de Janeiro, RJ, Brasil
| | | | | |
Collapse
|
35
|
Dendritic cells in systemic lupus erythematosus: From pathogenesis to therapeutic applications. J Autoimmun 2022; 132:102856. [DOI: 10.1016/j.jaut.2022.102856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
|
36
|
Shanina E, Kuhaudomlarp S, Siebs E, Fuchsberger FF, Denis M, da Silva Figueiredo Celestino Gomes P, Clausen MH, Seeberger PH, Rognan D, Titz A, Imberty A, Rademacher C. Targeting undruggable carbohydrate recognition sites through focused fragment library design. Commun Chem 2022; 5:64. [PMID: 36697615 PMCID: PMC9814205 DOI: 10.1038/s42004-022-00679-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/29/2022] [Indexed: 01/28/2023] Open
Abstract
Carbohydrate-protein interactions are key for cell-cell and host-pathogen recognition and thus, emerged as viable therapeutic targets. However, their hydrophilic nature poses major limitations to the conventional development of drug-like inhibitors. To address this shortcoming, four fragment libraries were screened to identify metal-binding pharmacophores (MBPs) as novel scaffolds for inhibition of Ca2+-dependent carbohydrate-protein interactions. Here, we show the effect of MBPs on the clinically relevant lectins DC-SIGN, Langerin, LecA and LecB. Detailed structural and biochemical investigations revealed the specificity of MBPs for different Ca2+-dependent lectins. Exploring the structure-activity relationships of several fragments uncovered the functional groups in the MBPs suitable for modification to further improve lectin binding and selectivity. Selected inhibitors bound efficiently to DC-SIGN-expressing cells. Altogether, the discovery of MBPs as a promising class of Ca2+-dependent lectin inhibitors creates a foundation for fragment-based ligand design for future drug discovery campaigns.
Collapse
Affiliation(s)
- Elena Shanina
- grid.419564.b0000 0004 0491 9719Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Mühlenberg 1, 14424 Potsdam, Germany ,grid.14095.390000 0000 9116 4836Freie Universität Berlin, Department of Chemistry and Biochemistry, Arnimallee 22, 14195 Berlin, Germany
| | - Sakonwan Kuhaudomlarp
- grid.450307.50000 0001 0944 2786University Grenoble Alpes, CNRS, CERMAV, Grenoble, France ,grid.10223.320000 0004 1937 0490Department of Biochemistry, Faculty of Science, Mahidol University, 10400 Bangkok, Thailand ,grid.10223.320000 0004 1937 0490Center for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, 10400 Bangkok, Thailand
| | - Eike Siebs
- grid.461899.bChemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Saarland University, Department of Chemistry, 66123 Saarbrücken, Germany ,grid.452463.2German Center for Infection Research (DZIF), Hannover-Braunschweig, Germany
| | - Felix F. Fuchsberger
- grid.419564.b0000 0004 0491 9719Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Mühlenberg 1, 14424 Potsdam, Germany ,grid.14095.390000 0000 9116 4836Freie Universität Berlin, Department of Chemistry and Biochemistry, Arnimallee 22, 14195 Berlin, Germany ,grid.10420.370000 0001 2286 1424University of Vienna, Department of Pharmaceutical Sciences, Althanstrasse 14, 1090 Vienna, Austria ,grid.10420.370000 0001 2286 1424University of Vienna, Department of Microbiology, Immunology and Genetics, Max F. Berutz Labs, Biocenter 5, 1030 Vienna, Austria
| | - Maxime Denis
- grid.10420.370000 0001 2286 1424University of Vienna, Department of Pharmaceutical Sciences, Althanstrasse 14, 1090 Vienna, Austria ,grid.10420.370000 0001 2286 1424University of Vienna, Department of Microbiology, Immunology and Genetics, Max F. Berutz Labs, Biocenter 5, 1030 Vienna, Austria
| | - Priscila da Silva Figueiredo Celestino Gomes
- grid.503326.10000 0004 0367 4780Laboratoire d’Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, 67400 Illkirch, France ,grid.252546.20000 0001 2297 8753Department of Physics, College of Sciences and Mathematics, Auburn University, 36849 Auburn, AL USA
| | - Mads H. Clausen
- grid.5170.30000 0001 2181 8870Technical University of Denmark, Center for Nanomedicine and Theranostics, Department of Chemistry, Kemitorvet 207, 2800 Kongens Lyngby, Denmark
| | - Peter H. Seeberger
- grid.419564.b0000 0004 0491 9719Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Mühlenberg 1, 14424 Potsdam, Germany ,grid.14095.390000 0000 9116 4836Freie Universität Berlin, Department of Chemistry and Biochemistry, Arnimallee 22, 14195 Berlin, Germany
| | - Didier Rognan
- grid.503326.10000 0004 0367 4780Laboratoire d’Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, 67400 Illkirch, France
| | - Alexander Titz
- grid.461899.bChemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Saarland University, Department of Chemistry, 66123 Saarbrücken, Germany ,grid.452463.2German Center for Infection Research (DZIF), Hannover-Braunschweig, Germany
| | - Anne Imberty
- grid.450307.50000 0001 0944 2786University Grenoble Alpes, CNRS, CERMAV, Grenoble, France
| | - Christoph Rademacher
- grid.419564.b0000 0004 0491 9719Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Mühlenberg 1, 14424 Potsdam, Germany ,grid.14095.390000 0000 9116 4836Freie Universität Berlin, Department of Chemistry and Biochemistry, Arnimallee 22, 14195 Berlin, Germany ,grid.10420.370000 0001 2286 1424University of Vienna, Department of Pharmaceutical Sciences, Althanstrasse 14, 1090 Vienna, Austria ,grid.10420.370000 0001 2286 1424University of Vienna, Department of Microbiology, Immunology and Genetics, Max F. Berutz Labs, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
37
|
Mazumdar V, Joshi K, Nandi BR, Namani S, Gupta VK, Radhakrishnan G. Host F-Box Protein 22 Enhances the Uptake of Brucella by Macrophages and Drives a Sustained Release of Proinflammatory Cytokines through Degradation of the Anti-Inflammatory Effector Proteins of Brucella. Infect Immun 2022; 90:e0006022. [PMID: 35420446 PMCID: PMC9119127 DOI: 10.1128/iai.00060-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 11/20/2022] Open
Abstract
Brucella species are intracellular bacterial pathogens, causing the worldwide zoonotic disease brucellosis. Brucella invades professional and nonprofessional phagocytic cells, followed by resisting intracellular killing and establishing a replication permissive niche. Brucella also modulates the innate and adaptive immune responses of the host for its chronic persistence. The complex intracellular cycle of Brucella depends in a major way on multiple host factors, but limited information is available on host and bacterial proteins that play an essential role in the invasion, intracellular replication, and modulation of host immune responses. By employing a small interfering RNA (siRNA) screening, we identified a role for the host protein FBXO22 in the Brucella-macrophage interaction. FBXO22 is the key element in the SCF E3 ubiquitination complex, where it determines the substrate specificity for ubiquitination and degradation of various host proteins. Downregulation of FBXO22 by siRNA or the CRISPR-Cas9 system resulted in diminished uptake of Brucella into macrophages, which was dependent on NF-κB-mediated regulation of phagocytic receptors. FBXO22 expression was upregulated in Brucella-infected macrophages, which resulted in induction of phagocytic receptors and enhanced production of proinflammatory cytokines through NF-κB. Furthermore, we found that FBXO22 recruits the effector proteins of Brucella, including the anti-inflammatory proteins TcpB and OMP25, for degradation through the SCF complex. We did not observe any role for another F-box-containing protein of the SCF complex, β-TrCP, in the Brucella-macrophage interaction. Our findings unravel novel functions of FBXO22 in host-pathogen interaction and its contribution to pathogenesis of infectious diseases.
Collapse
Affiliation(s)
- Varadendra Mazumdar
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Binita Roy Nandi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Swapna Namani
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Vivek Kumar Gupta
- ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, India
| | - Girish Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| |
Collapse
|
38
|
Alves I, Fernandes Â, Santos-Pereira B, Azevedo CM, Pinho SS. Glycans as a key factor in self and non-self discrimination: Impact on the breach of immune tolerance. FEBS Lett 2022; 596:1485-1502. [PMID: 35383918 DOI: 10.1002/1873-3468.14347] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Glycans are carbohydrates that are made by all organisms and covalently conjugated to other biomolecules. Glycans cover the surface of both human cells and pathogens and are fundamental to defining the identity of a cell or an organism, thereby contributing to discriminating self from non-self. As such, glycans are a class of "Self-Associated Molecular Patterns" that can fine-tune host inflammatory processes. In fact, glycans can be sensed and recognized by a variety of glycan-binding proteins (GBP) expressed by immune cells, such as galectins, siglecs and C-type lectins, which recognize changes in the cellular glycosylation, instructing both pro-inflammatory or anti-inflammatory responses. In this review, we introduce glycans as cell-identification structures, discussing how glycans modulate host-pathogen interactions and how they can fine-tune inflammatory processes associated with infection, inflammation and autoimmunity. Finally, from the clinical standpoint, we discuss how glycoscience research can benefit life sciences and clinical medicine by providing a source of valuable biomarkers and therapeutic targets for immunity.
Collapse
Affiliation(s)
- Inês Alves
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ângela Fernandes
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Beatriz Santos-Pereira
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Catarina M Azevedo
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Salomé S Pinho
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| |
Collapse
|
39
|
Firdaus FZ, Skwarczynski M, Toth I. Developments in Vaccine Adjuvants. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:145-178. [PMID: 34918245 DOI: 10.1007/978-1-0716-1892-9_8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccines, including subunit, recombinant, and conjugate vaccines, require the use of an immunostimulator/adjuvant for maximum efficacy. Adjuvants not only enhance the strength and longevity of immune responses but may also influence the type of response. In this chapter, we review the adjuvants that are available for use in human vaccines, such as alum, MF59, AS03, and AS01. We extensively discuss their composition, characteristics, mechanism of action, and effects on the immune system. Additionally, we summarize recent trends in adjuvant discovery, providing a brief overview of saponins, TLRs agonists, polysaccharides, nanoparticles, cytokines, and mucosal adjuvants.
Collapse
Affiliation(s)
- Farrhana Ziana Firdaus
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,Institute of Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
40
|
Cramer J. Medicinal chemistry of the myeloid C-type lectin receptors Mincle, Langerin, and DC-SIGN. RSC Med Chem 2021; 12:1985-2000. [PMID: 35024612 PMCID: PMC8672822 DOI: 10.1039/d1md00238d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
In their role as pattern-recognition receptors on cells of the innate immune system, myeloid C-type lectin receptors (CLRs) assume important biological functions related to immunity, homeostasis, and cancer. As such, this family of receptors represents an appealing target for therapeutic interventions for modulating the outcome of many pathological processes, in particular related to infectious diseases. This review summarizes the current state of research into glycomimetic or drug-like small molecule ligands for the CLRs Mincle, Langerin, and DC-SIGN, which have potential therapeutic applications in vaccine research and anti-infective therapy.
Collapse
Affiliation(s)
- Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University of Düsseldorf Universitätsstr. 1 40225 Düsseldorf Germany
| |
Collapse
|
41
|
Elemam NM, Ramakrishnan RK, Hundt JE, Halwani R, Maghazachi AA, Hamid Q. Innate Lymphoid Cells and Natural Killer Cells in Bacterial Infections: Function, Dysregulation, and Therapeutic Targets. Front Cell Infect Microbiol 2021; 11:733564. [PMID: 34804991 PMCID: PMC8602108 DOI: 10.3389/fcimb.2021.733564] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Infectious diseases represent one of the largest medical challenges worldwide. Bacterial infections, in particular, remain a pertinent health challenge and burden. Moreover, such infections increase over time due to the continuous use of various antibiotics without medical need, thus leading to several side effects and bacterial resistance. Our innate immune system represents our first line of defense against any foreign pathogens. This system comprises the innate lymphoid cells (ILCs), including natural killer (NK) cells that are critical players in establishing homeostasis and immunity against infections. ILCs are a group of functionally heterogenous but potent innate immune effector cells that constitute tissue-resident sentinels against intracellular and extracellular bacterial infections. Being a nascent subset of innate lymphocytes, their role in bacterial infections is not clearly understood. Furthermore, these pathogens have developed methods to evade the host immune system, and hence permit infection spread and tissue damage. In this review, we highlight the role of the different ILC populations in various bacterial infections and the possible ways of immune evasion. Additionally, potential immunotherapies to manipulate ILC responses will be briefly discussed.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rakhee K Ramakrishnan
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jennifer E Hundt
- Lübeck Institute for Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Rabih Halwani
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Prince Abdullah Ben Khaled Celiac Disease Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Azzam A Maghazachi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| |
Collapse
|
42
|
Willment JA. Fc-conjugated C-type lectin receptors: Tools for understanding host-pathogen interactions. Mol Microbiol 2021; 117:632-660. [PMID: 34709692 DOI: 10.1111/mmi.14837] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022]
Abstract
The use of soluble fusion proteins of pattern recognition receptors (PRRs) used in the detection of exogenous and endogenous ligands has helped resolve the roles of PRRs in the innate immune response to pathogens, how they shape the adaptive immune response, and function in maintaining homeostasis. Using the immunoglobulin (Ig) crystallizable fragment (Fc) domain as a fusion partner, the PRR fusion proteins are soluble, stable, easily purified, have increased affinity due to the Fc homodimerization properties, and consequently have been used in a wide range of applications such as flow cytometry, screening of protein and glycan arrays, and immunofluorescent microscopy. This review will predominantly focus on the recognition of pathogens by the cell membrane-expressed glycan-binding proteins of the C-type lectin receptor (CLR) subgroup of PRRs. PRRs bind to conserved pathogen-associated molecular patterns (PAMPs), such as glycans, usually located within or on the outer surface of the pathogen. Significantly, many glycans structures are identical on both host and pathogen (e.g. the Lewis (Le) X glycan), allowing the use of Fc CLR fusion proteins with known endogenous and/or exogenous ligands as tools to identify pathogen structures that are able to interact with the immune system. Screens of highly purified pathogen-derived cell wall components have enabled identification of many unique PAMP structures recognized by CLRs. This review highlights studies using Fc CLR fusion proteins, with emphasis on the PAMPs found in fungi, bacteria, viruses, and parasites. The structure and unique features of the different CLR families is presented using examples from a broad range of microbes whenever possible.
Collapse
Affiliation(s)
- Janet A Willment
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
43
|
Kalra R, Tiwari D, Dkhar HK, Bhagyaraj E, Kumar R, Bhardwaj A, Gupta P. Host factors subverted by Mycobacterium tuberculosis: Potential targets for host directed therapy. Int Rev Immunol 2021; 42:43-70. [PMID: 34678117 DOI: 10.1080/08830185.2021.1990277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Despite new approaches in the diagnosis and treatment of tuberculosis (TB), it continues to be a major health burden. Several immunotherapies that potentiate the immune response have come up as adjuncts to drug therapies against drug resistant TB strains; however, there needs to be an urgent appraisal of host specific drug targets for improving their clinical management and to curtail disease progression. Presently, various host directed therapies (HDTs) exist (repurposed drugs, nutraceuticals, monoclonal antibodies and immunomodulatory agents), but these mostly address molecules that combat disease progression. AREAS COVERED The current review discusses major Mycobacterium tuberculosis (M. tuberculosis) survival paradigms inside the host and presents a plethora of host targets subverted by M. tuberculosis which can be further explored for future HDTs. The host factors unique to M. tuberculosis infection (in humans) have also been identified through an in-silico interaction mapping. EXPERT OPINION HDTs could become the next-generation adjunct therapies in order to counter antimicrobial resistance and virulence, as well as to reduce the duration of existing TB treatments. However, current scientific efforts are largely directed toward combatants rather than host molecules co-opted by M. tuberculosis for its survival. This might drive the immune system to a hyper-inflammatory condition; therefore, we emphasize that host factors subverted by M. tuberculosis, and their subsequent neutralization, must be considered for development of better HDTs.
Collapse
Affiliation(s)
- Rashi Kalra
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Drishti Tiwari
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Hedwin Kitdorlang Dkhar
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Ella Bhagyaraj
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Rakesh Kumar
- Bioinformatics Center, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Anshu Bhardwaj
- Bioinformatics Center, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Pawan Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
44
|
van der Vlist M, Ramos MIP, van den Hoogen LL, Hiddingh S, Timmerman LM, de Hond TAP, Kaan ED, van der Kroef M, Lebbink RJ, Peters FMA, Khoury-Hanold W, Fritsch-Stork R, Radstake TRDJ, Meyaard L. Signaling by the inhibitory receptor CD200R is rewired by type I interferon. Sci Signal 2021; 14:eabb4324. [PMID: 34637328 DOI: 10.1126/scisignal.abb4324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CD200 receptor 1 (CD200R) is an inhibitory immunoreceptor that suppresses Toll-like receptor (TLR)–induced cytokine production through the adaptor protein Dok2 and the GTPase activating protein (GAP) p120-RasGAP, which can be cleaved during mild cellular stress. We found that in the presence of cleaved p120-RasGAP, CD200R lost its capacity to inhibit phosphorylation of ribosomal S6 protein (rpS6), suggesting the reduced activity of mammalian target of rapamycin complex 1 (mTORC1). Furthermore, treatment of human peripheral blood mononuclear cells (PBMC) with interferon-α (IFN-α) resulted in increased amounts of cleaved p120-RasGAP. Upon pretreatment of cells with increasing concentrations of IFN-α, CD200R switched from inhibiting to potentiating the TLR7- and TLR8-induced expression of the gene encoding IFN-γ, a cytokine that is important for innate and adaptive immunity and is implicated in systemic lupus erythematosus (SLE) pathogenesis. PBMC from patients with SLE, a prototypic type I IFN disease, had an increased abundance of cleaved p120-RasGAP compared to that in cells from healthy controls. In a subset of SLE patients, CD200R stopped functioning as an inhibitory receptor or potentiated TLR-induced IFNG mRNA expression. Thus, our data suggest that type I IFN rewires CD200R signaling to be proinflammatory, which could contribute to the perpetuation of inflammation in patients with SLE.
Collapse
Affiliation(s)
- Michiel van der Vlist
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - M Inês Pascoal Ramos
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Lucas L van den Hoogen
- Center for Translational Immunology, Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sanne Hiddingh
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Laura M Timmerman
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Titus A P de Hond
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Ellen D Kaan
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Maarten van der Kroef
- Center for Translational Immunology, Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Florence M A Peters
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - William Khoury-Hanold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruth Fritsch-Stork
- Center for Translational Immunology, Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Timothy R D J Radstake
- Center for Translational Immunology, Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
45
|
Yan Z, Wang H, Mu L, Hu ZD, Zheng WQ. Regulatory roles of extracellular vesicles in immune responses against Mycobacterium tuberculosis infection. World J Clin Cases 2021; 9:7311-7318. [PMID: 34616797 PMCID: PMC8464473 DOI: 10.12998/wjcc.v9.i25.7311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/19/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are cystic vesicles naturally released by most mammalian cells and bacteria. EV contents include proteins, lipids, and nucleic acids. EVs can act as messengers to transmit a variety of molecules to recipient cells and thus play important regulatory roles in intercellular signal transduction. EVs, released by either a host cell or a pathogen, can carry pathogen-associated antigens and thus act as modulators of immune responses. EVs derived from Mycobacterium tuberculosis (Mtb)-infected cells can regulate the innate immune response through various pathways, such as regulating the release of inflammatory cytokines. In addition, EVs can mediate antigen presentation and regulate the adaptive immune response by transmitting immunoregulatory molecules to T helper cells. In this review, we summarize the regulatory roles of EVs in the immune response against Mtb.
Collapse
Affiliation(s)
- Zhi Yan
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Hua Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Lan Mu
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Zhi-De Hu
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Wen-Qi Zheng
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
- Department of Parasitology, the College of Basic Medical Sciences of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| |
Collapse
|
46
|
Di Lorenzo F, Duda KA, Lanzetta R, Silipo A, De Castro C, Molinaro A. A Journey from Structure to Function of Bacterial Lipopolysaccharides. Chem Rev 2021; 122:15767-15821. [PMID: 34286971 DOI: 10.1021/acs.chemrev.0c01321] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Lipopolysaccharide (LPS) is a crucial constituent of the outer membrane of most Gram-negative bacteria, playing a fundamental role in the protection of bacteria from environmental stress factors, in drug resistance, in pathogenesis, and in symbiosis. During the last decades, LPS has been thoroughly dissected, and massive information on this fascinating biomolecule is now available. In this Review, we will give the reader a third millennium update of the current knowledge of LPS with key information on the inherent peculiar carbohydrate chemistry due to often puzzling sugar residues that are uniquely found on it. Then, we will drive the reader through the complex and multifarious immunological outcomes that any given LPS can raise, which is strictly dependent on its chemical structure. Further, we will argue about issues that still remain unresolved and that would represent the immediate future of LPS research. It is critical to address these points to complete our notions on LPS chemistry, functions, and roles, in turn leading to innovative ways to manipulate the processes involving such a still controversial and intriguing biomolecule.
Collapse
Affiliation(s)
- Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy
| | - Katarzyna A Duda
- Research Center Borstel Leibniz Lung Center, Parkallee 4a, 23845 Borstel, Germany
| | - Rosa Lanzetta
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy
| | - Cristina De Castro
- Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Department of Agricultural Sciences, University of Naples Federico II, Via Università 96, 80055 Portici, Naples, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
47
|
Holzheimer M, Buter J, Minnaard AJ. Chemical Synthesis of Cell Wall Constituents of Mycobacterium tuberculosis. Chem Rev 2021; 121:9554-9643. [PMID: 34190544 PMCID: PMC8361437 DOI: 10.1021/acs.chemrev.1c00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The pathogen Mycobacterium tuberculosis (Mtb), causing
tuberculosis disease, features an extraordinary
thick cell envelope, rich in Mtb-specific lipids,
glycolipids, and glycans. These cell wall components are often directly
involved in host–pathogen interaction and recognition, intracellular
survival, and virulence. For decades, these mycobacterial natural
products have been of great interest for immunology and synthetic
chemistry alike, due to their complex molecular structure and the
biological functions arising from it. The synthesis of many of these
constituents has been achieved and aided the elucidation of their
function by utilizing the synthetic material to study Mtb immunology. This review summarizes the synthetic efforts of a quarter
century of total synthesis and highlights how the synthesis layed
the foundation for immunological studies as well as drove the field
of organic synthesis and catalysis to efficiently access these complex
natural products.
Collapse
Affiliation(s)
- Mira Holzheimer
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Jeffrey Buter
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| |
Collapse
|
48
|
Identification and expression analysis of group II C-type lectin domain containing receptors in grass carp Ctenopharyngodon idella. Gene 2021; 789:145668. [PMID: 33882323 DOI: 10.1016/j.gene.2021.145668] [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: 12/15/2020] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 01/16/2023]
Abstract
Group II C-type lectin domain (CTLD) containing receptors belong to a large family of pattern recognition receptors which mainly act on the innate immunity. They are structurally related and consist of a cytoplasmic immunoreceptor tyrosine-based inhibitory motif (ITIM) and a single extracellular CTLD. Although they have been described in teleost fish, their involvement in immune responses is not well understood. In this study, four immune-related lectin-like receptors (termed CiILLR1 and CiILLR5-7), belonging to the group II CTLD receptors, were identified in grass carp (Ctenopharyngodon idella). They contain a short cytoplasmic tail and a single CTLD in the extracellular region. The CiILLR1 has a WxHxxxxxY motif similar to the WxHxxxxY motif which is required for the recognition of β-glucans by some of the group II CTLD containing lectins in mammals. Further, a modified QPD motif (EPD) known to be involved in binding to carbohydrate ligands is present in the CiILLR1, 5 and 6. However, CiILLR7 lacks these motifs. Expression analysis revealed that they were constitutively expressed in the head kidney and spleen. Moreover, CiILLR1, 5 and 6 could be up-regulated in the head kidney and spleen of fish after infection with Flavobacterium columnare and in the primary head kidney leukocytes by LPS and PHA. Expression of CiILLR1, CiILLR5 and CiILLR6 were mainly detected in the enriched lymphocytes whilst CiILLR7 was expressed in the enriched monocytes/macrophages. The results expand existing knowledge on the immune responses of the C-type lectin receptors in teleost fish.
Collapse
|
49
|
In silico screening and identification of deleterious missense SNPs along with their effects on CD-209 gene: An insight to CD-209 related-diseases. PLoS One 2021; 16:e0247249. [PMID: 33635927 PMCID: PMC7909662 DOI: 10.1371/journal.pone.0247249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
DC-SIGN receptor articulated by macrophages and dendritic cells is encoded by CD209 gene and plays a role to activate and proliferate the T-lymphocytes in response of virus attack. The dysfunctional activity of DC-SIGN receptor because of missense SNPs can lead to cause dengue haemorrhage fever, HIV-1 infection etc. Out of 11 transcripts of CD209, all missense SNPs of canonical transcript were retrieved from Ensembl database and evaluated by their deleteriousness by using Polyphen-2, PMut, SIFT, MutPred, PROVEAN and PhD-SNP together with stimulation of its complete 3D structure. 10 nsSNPs were chosen depending on both the significance value of nsSNP and their prediction among SNPs evaluating servers which are based on different algorithms. Moreover, the position and native role of 10 nsSNPs in wild 3D model has been described which assist to acknowledge their importance. This study urges the researcher’s community to experimentally validate these SNPs and their association in causing the diseases like dengue fever, Tuberculosis etc.
Collapse
|
50
|
Maphasa RE, Meyer M, Dube A. The Macrophage Response to Mycobacterium tuberculosis and Opportunities for Autophagy Inducing Nanomedicines for Tuberculosis Therapy. Front Cell Infect Microbiol 2021; 10:618414. [PMID: 33628745 PMCID: PMC7897680 DOI: 10.3389/fcimb.2020.618414] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
The major causative agent of tuberculosis (TB), i.e., Mycobacterium tuberculosis (Mtb), has developed mechanisms to evade host defense responses and persist within host cells for prolonged periods of time. Mtb is also increasingly resistant to existing anti-TB drugs. There is therefore an urgent need to develop new therapeutics for TB and host directed therapies (HDTs) hold potential as effective therapeutics for TB. There is growing interest in the induction of autophagy in Mtb host cells using autophagy inducing compounds (AICs). Nanoparticles (NPs) can enhance the effect of AICs, thus improving stability, enabling cell targeting and providing opportunities for multimodal therapy. In this review, we focus on the macrophage responses to Mtb infection, in particular, the mechanistic aspects of autophagy and the evasion of autophagy by intracellular Mtb. Due to the overlap between the onset of autophagy and apoptosis; we also focus on the relationship between apoptosis and autophagy. We will also review known AICs in the context of Mtb infection. Finally, we discuss the applications of NPs in inducing autophagy with the intention of sharing insights to encourage further research and development of nanomedicine HDTs for TB therapy.
Collapse
Affiliation(s)
- Retsepile E Maphasa
- Infectious Disease Nanomedicine Research Group, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| | - Mervin Meyer
- DST/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Cape Town, South Africa
| | - Admire Dube
- Infectious Disease Nanomedicine Research Group, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| |
Collapse
|