1
|
D'Angelo E, Rampado R, Sensi F, Marangio A, Rossi AD, Repetto O, Steffan A, Corallo D, Aveic S, Bianchi G, Collino F, Caliceti P, Spolverato G, Agostini M. Tumor microenvironment-mimicking macrophage nanovesicles as a targeted therapy platform for colorectal cancer. Int J Pharm 2025; 670:125169. [PMID: 39756598 DOI: 10.1016/j.ijpharm.2025.125169] [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: 10/07/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Macrophages are a pivotal immune cell population in the tumor microenvironment of colorectal cancer (CRC). Differently-polarized macrophages could be exploited to yield naturally-tailored biomimetic nanoparticles for CRC targeting. Here, membrane proteins were isolated from the THP-1 cell line, and anti-tumor macrophages (M1) were obtained from differentiation of THP-1. Membrane proteins were isolated from THP-1 and M1 and used to produce lipid nanovesicles (LNVs; T-LNVs and M1-LNVs) by microfluidic process, which were loaded with doxorubicin (DOXO). The DOXO loaded T-LNVs and M1-LNVs showed similar size (120-145 nm), PDI (0.11-0.28), zeta potential (-15 to -30 mV) and drug loading efficiency (65-75 %). Mass-spectrometry confirmed the presence of the membrane proteins in the LNVs. The abundance of proteins related to stealth properties, cancer targeting, endothelial adhesion and immune-related markers was significantly different in T-LNVs and M1-LNVs. Cell culture studies showed that M1-LNVs possessed higher cancer cell targeting, uptake and cytotoxicity compared to T-LNVs. In vivo studies performed with zebrafish embryos showed that M1-LNVs yielded higher cancer cell targeting and cytotoxicity while systemic cytotoxicity was lower compared to free DOXO. These findings confirm the potentiality and versatility of M1-LNVs for cancer treatment, which could be exploited as new avenue of nanoparticles-based therapies for precision medicine.
Collapse
Affiliation(s)
- Edoardo D'Angelo
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padua, Italy; NanoInspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.
| | - Riccardo Rampado
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer, Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel; Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Francesca Sensi
- NanoInspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy; Department of Women and Children's Health, University of Padova, via Giustiniani 2, 35128 Padua, Italy
| | - Asia Marangio
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padua, Italy
| | - Anna De Rossi
- Department of Women and Children's Health, University of Padova, via Giustiniani 2, 35128 Padua, Italy
| | - Ombretta Repetto
- Immunopathology and Cancer Biomarkers, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Gaia Bianchi
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Dipartimento di Eccellenza 2023-2027, University of Milano, Milan, Italy
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Dipartimento di Eccellenza 2023-2027, University of Milano, Milan, Italy; Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milano, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Gaya Spolverato
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padua, Italy
| | - Marco Agostini
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padua, Italy; NanoInspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| |
Collapse
|
2
|
Lundstrom K. Immunobiology and immunotherapy of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:73-133. [PMID: 40246352 DOI: 10.1016/bs.pmbts.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The SARS-CoV-2 outbreak in late 2019 triggered a major increase in activities related to immunobiology and immunotherapy to cope with and find solutions to end the COVID-19 pandemic. The unprecedented approach to research and development of drugs and vaccines against SARS-CoV-2 has substantially improved the understanding of immunobiology for COVID-19, which can also be applied to other infectious diseases. Major efforts were dedicated to the repurposing of existing antiviral drugs and the development of novel ones. For this reason, numerous approaches to evaluating interferons, immunoglobulins, and cytokine inhibitors have been conducted. Antibody-based therapies, especially employing monoclonal antibodies have also been on the agenda. Cell-based therapies involving dendritic cells, macrophages, and CAR T-cell approaches have been evaluated. Many existing antiviral drugs have been repurposed for COVID-19 and novel formulations have been tested. The extraordinarily rapid development of efficient vaccines led to the breakthrough of novel vaccine approaches such as mRNA-based vaccines saving millions of lives. Waning immunity of existing vaccines and emerging SARS-CoV-2 variants have required additional booster vaccinations and re-engineering of new versions of COVID-19 vaccines.
Collapse
|
3
|
Hage SF, Bi DE, Kinkade S, Vera Cruz D, Srinath A, Jhaveri A, Romanos S, Bindal A, Lightle R, Little JC, Shenkar R, Alcazar-Felix RJ, Lee J, Stadnik A, Sidebottom A, Carroll TJ, Ji Y, Koskimaki J, Polster SP, Girard R, Awad IA. Circulating molecules reflect imaging biomarkers of hemorrhage in cerebral cavernous malformations. J Cereb Blood Flow Metab 2025:271678X251314366. [PMID: 39829356 PMCID: PMC11748132 DOI: 10.1177/0271678x251314366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Increases in mean lesional iron content by quantitative susceptibility mapping (QSM) by ≥6% and/or vascular permeability by dynamic contrast enhanced quantitative perfusion (DCEQP) by ≥40% on MRI have been associated with new symptomatic hemorrhage (SH) in cerebral cavernous malformations (CCMs). It is not known if plasma biomarkers can reflect these changes within the lesion proper. This cohort study enrolled 46 CCM patients with SH in the prior year. Plasma samples, QSM and DCEQP were simultaneously acquired at the beginning and end of 60 one-year epochs of prospective follow-up. Plasma levels of 16 proteins and 12 metabolites linked to CCM hemorrhage were assessed by enzyme-linked immunosorbent assay and liquid-chromatography mass spectrometry, respectively. A weighted model combining the percent changes in plasma levels in roundabout guidance receptor-4, cluster of differentiation 14, thrombomodulin and acetyl-L-carnitine reflected a mean increase in QSM ≥ 6% (97.2% and 100% specificity/sensitivity, p = 3.1 × 10-13). A weighted combination of percent changes in plasma levels of endoglin, pipecolic acid, arachidonic acid and hypoxanthine correlated with an increase in mean DCEQP ≥40% (99.6% specificity and 100% sensitivity, p = 4.1 × 10-17). This is a first report linking with great accuracy changes of circulating molecules to imaging changes reflecting new SH during prospective follow-up of CCMs.
Collapse
Affiliation(s)
- Stephanie F Hage
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Dehua E Bi
- Department of Public Health Sciences, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Serena Kinkade
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Diana Vera Cruz
- Center for Research Informatics, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Aditya Jhaveri
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Akash Bindal
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Jessica C Little
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Roberto J Alcazar-Felix
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Justine Lee
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Ashley Sidebottom
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Timothy J Carroll
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Yuan Ji
- Department of Public Health Sciences, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Janne Koskimaki
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| |
Collapse
|
4
|
Boliukh I, Rombel-Bryzek A, Bułdak RJ. Lectins in oncology and virology: Mechanisms of anticancer activity and SARS-CoV-2 inhibition. Int J Biol Macromol 2024; 275:133664. [PMID: 38969035 DOI: 10.1016/j.ijbiomac.2024.133664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Lectins are proteins or glycoproteins of non-immune origin with carbohydrate-binding properties. They are found both prokaryotic and eukaryotic organisms. The most abundant source of the lectins are plants. Many lectins have anticancer effects by directly exerting cytotoxic effects on malignant cells or indirectly activating the immune system. Lectins also have antiviral activities. These proteins can recognise glycoproteins on the surface of enveloped viruses and bind to them. This creates a physical barrier between them and the corresponding receptors on the surface of the host cell, which prevents the virus from entering the cell and can thus effectively inhibit the replication of the virus. In this review, we focus on the anticancer activities of selected lectins and the underlying mechanisms. We also discuss different types of lectins with antiviral activity. We have paid special attention to lectins with inhibitory activity against SARS-CoV-2. Finally, we outline the challenges of using lectins in therapy and suggest future research directions.
Collapse
Affiliation(s)
- Iryna Boliukh
- Institute of Medical Sciences, University of Opole, Opole, Poland
| | | | - Rafał J Bułdak
- Institute of Medical Sciences, University of Opole, Opole, Poland
| |
Collapse
|
5
|
Habanjar O, Nehme R, Goncalves-Mendes N, Cueff G, Blavignac C, Aoun J, Decombat C, Auxenfans C, Diab-Assaf M, Caldefie-Chézet F, Delort L. The obese inflammatory microenvironment may promote breast DCIS progression. Front Immunol 2024; 15:1384354. [PMID: 39072314 PMCID: PMC11272476 DOI: 10.3389/fimmu.2024.1384354] [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: 02/09/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Ductal carcinoma in situ (DCIS), characterized by a proliferation of neoplastic cells confined within the mammary ducts, is distinctly isolated from the surrounding stroma by an almost uninterrupted layer of myoepithelial cells (MECs) and by the basement membrane. Heightened interactions within the adipose microenvironment, particularly in obese patients, may play a key role in the transition from DCIS to invasive ductal carcinoma (IDC), which is attracting growing interest in scientific research. Adipose tissue undergoes metabolic changes in obesity, impacting adipokine secretion and promoting chronic inflammation. This study aimed to assess the interactions between DCIS, including in situ cancer cells and MECs, and the various components of its inflammatory adipose microenvironment (adipocytes and macrophages). Methods To this end, a 3D co-culture model was developed using bicellular bi-fluorescent DCIS-like tumoroids, adipose cells, and macrophages to investigate the influence of the inflammatory adipose microenvironment on DCIS progression. Results The 3D co-culture model demonstrated an inhibition of the expression of genes involved in apoptosis (BAX, BAG1, BCL2, CASP3, CASP8, and CASP9), and an increase in genes related to cell survival (TP53, JUN, and TGFB1), inflammation (TNF-α, PTGS2, IL-6R), invasion and metastasis (TIMP1 and MMP-9) in cancer cells of the tumoroids under inflammatory conditions versus a non-inflammatory microenvironment. On the contrary, it confirmed the compromised functionality of MECs, resulting in the loss of their protective effects against cancer cells. Adipocytes from obese women showed a significant increase in the expression of all studied myofibroblast-associated genes (myoCAFs), such as FAP and α-SMA. In contrast, adipocytes from normal-weight women expressed markers of inflammatory fibroblast phenotypes (iCAF) characterized by a significant increase in the expression of LIF and inflammatory cytokines such as TNF-α, IL-1β, IL-8, and CXCL-10. These changes also influenced macrophage polarization, leading to a pro-inflammatory M1 phenotype. In contrast, myoCAF-associated adipocytes, and the cancer-promoting microenvironment polarized macrophages towards an M2 phenotype, characterized by high CD163 receptor expression and IL-10 and TGF-β secretion. Discussion Reciprocal interactions between the tumoroid and its microenvironment, particularly in obesity, led to transcriptomic changes in adipocytes and macrophages, may participate in breast cancer progression while disrupting the integrity of the MEC layer. These results underlined the importance of adipose tissue in cancer progression.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Rawan Nehme
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Gwendal Cueff
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Christelle Blavignac
- Université Clermont-Auvergne, Centre d’Imagerie Cellulaire Santé (CCIS), Clermont-Ferrand, France
| | - Jessy Aoun
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Céline Auxenfans
- Banque de tissus et de cellules, Hôpital Edouard-Herriot, Lyon, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Moléculaire et Pharmacologie Anticancéreuse, Faculté des Sciences II, Université libanaise Fanar, Beirut, Lebanon
| | | | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| |
Collapse
|
6
|
Martins Oliveira-Brito PK, de Campos GY, Guimarães JG, Machado MP, Serafim LC, Lazo Chica JE, Roque-Barreira MC, da Silva TA. Adjuvant ArtinM favored the host immunity against Cryptococcus gattii infection in C57BL/6 mice. Immunotherapy 2024; 16:733-748. [PMID: 38940276 PMCID: PMC11421300 DOI: 10.1080/1750743x.2024.2360384] [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: 01/15/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024] Open
Abstract
Aim: Cryptococcus gattii causes a severe fungal infection with high mortality rate among immunosuppressed and immunocompetent individuals. Due to limitation of current antifungal treatment, new immunotherapeutic approaches are explored.Methods: This study investigated an immunization strategy utilizing heat-inactivated C. gattii with ArtinM as an adjuvant. C57BL/6 mice were intranasally immunized with heat-killed C. gattii and ArtinM was administrated either before immunization or along with HK-C. gattii. Mice were infected with C. gattii and the efficacy of the immunization protocol was evaluated.Results: Mice that received ArtinM exhibited increased levels of IL-10 and relative expression of IL-23 in the lungs, reduced fungal burden and preserved tissue integrity post-infection.Conclusion: Adjuvant ArtinM improved immunization against C. gattii infection in C57BL/6 mice.
Collapse
Affiliation(s)
- Patrícia Kellen Martins Oliveira-Brito
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gabriela Yamazaki de Campos
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Júlia Garcia Guimarães
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Michele Procópio Machado
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Letícia Costa Serafim
- Microbiology Postgraduate Program of the Microbiology Department of the Biomedical Sciences Institute (ICB) of University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Javier Emílio Lazo Chica
- Institute of Natural & Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago Aparecido da Silva
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Clinical Hematology Lab, Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara (FCFAR), Sao Paulo State University (UNESP), Araraquara, São Paulo, Brazil
- National Institute of Science & Technology in Human Pathogenic Fungi, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto, São Paulo,Brazil
| |
Collapse
|
7
|
Tang J, Zhang S, Jiang L, Liu J, Xu J, Jiang C, Chen Z, Zhou X, Fuller C, Huang J, Chen H, Yang G, Bai C, Yin D, Li B, Chi H. Causal relationship between immune cells and hepatocellular carcinoma: a Mendelian randomisation study. J Cancer 2024; 15:4219-4231. [PMID: 38947379 PMCID: PMC11212088 DOI: 10.7150/jca.96744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Background: Hepatocellular carcinoma (HCC), the predominant malignancy of the digestive tract, ranks as the third most common cause of cancer-related mortality globally, significantly impeding human health and lifespan. Emerging immunotherapeutic approaches have ignited fresh optimism for patient outcomes. This investigation probes the link between 731 immune cell phenotypes and HCC through Mendelian Randomization and single-cell sequencing, aiming to unearth viable drug targets and dissect HCC's etiology. Methods: We conducted an exhaustive two-sample Mendelian Randomization analysis to ascertain the causal links between immune cell features and HCC, utilizing publicly accessible genetic datasets to explore the causal connections of 731 immune cell traits with HCC susceptibility. The integrity, diversity, and potential horizontal pleiotropy of these findings were rigorously assessed through extensive sensitivity analyses. Furthermore, single-cell sequencing was employed to penetrate the pathogenic underpinnings of HCC. Results: Establishing a significance threshold of pval_Inverse.variance.weighted at 0.05, our study pinpointed five immune characteristics potentially elevating HCC risk: B cell % CD3- lymphocyte (TBNK panel), CD25 on IgD+ (B cell panel), HVEM on TD CD4+ (Maturation stages of T cell panel), CD14 on CD14+ CD16- monocyte (Monocyte panel), CD4 on CD39+ activated Treg ( Treg panel). Conversely, various cellular phenotypes tied to BAFF-R expression emerged as protective elements. Single-cell sequencing unveiled profound immune cell phenotype interactions, highlighting marked disparities in cell communication and metabolic activities. Conclusion: Leveraging MR and scRNA-seq techniques, our study elucidates potential associations between 731 immune cell phenotypes and HCC, offering a window into the molecular interplays among cellular phenotypes, and addressing the limitations of mono-antibody therapeutic targets.
Collapse
Affiliation(s)
- Jingyi Tang
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital, Southwest Medical University, Sichuan, China
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Shengke Zhang
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lai Jiang
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jie Liu
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital, Southwest Medical University, Sichuan, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou 635000, China
| | - Jiayu Xu
- School of Science, Minzu University of China, Beijing, 100081 China
| | - Chenglu Jiang
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Zipei Chen
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xuancheng Zhou
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Claire Fuller
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jinbang Huang
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Haiqing Chen
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens 45701, OH, USA
| | - Changsong Bai
- Department of General Surgery, Xuyong People's Hospital, Luzhou, China
| | - Defeng Yin
- Department of Emergency Medicine, The Affiliated Hospital, Southwest Medical University, 646000 Luzhou, China
- Department of Emergency Medicine, Xuyong People's Hospital, Luzhou, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital, Southwest Medical University, Sichuan, China
| | - Hao Chi
- Department of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of General Surgery, Xuyong People's Hospital, Luzhou, China
| |
Collapse
|
8
|
Abouelkheir M, Roy T, Krzyscik MA, Özdemir E, Hristova K. Investigations of membrane protein interactions in cells using fluorescence microscopy. Curr Opin Struct Biol 2024; 86:102816. [PMID: 38648680 PMCID: PMC11141325 DOI: 10.1016/j.sbi.2024.102816] [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: 10/30/2023] [Revised: 03/06/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
The interactions between proteins in membranes govern many cellular functions. Our ability to probe for such interactions has greatly evolved in recent years due to the introduction of new fluorescence techniques. As a result, we currently have a choice of methods that can be used to assess the spatial distribution of a membrane protein, its association state, and the thermodynamic stability of the oligomers in the native milieu. These biophysical measurements have revealed new insights into important biological processes in cellular membranes.
Collapse
Affiliation(s)
- Mahmoud Abouelkheir
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA; Chemistry-Biology Interface Program, Johns Hopkins University, Baltimore MD 21218, USA
| | - Tanaya Roy
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Mateusz A Krzyscik
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Ece Özdemir
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA
| | - Kalina Hristova
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore MD 21218, USA; Chemistry-Biology Interface Program, Johns Hopkins University, Baltimore MD 21218, USA.
| |
Collapse
|
9
|
Gupta KH, Giurini EF, Zloza A. Seasonal influenza vaccines differentially activate and modulate toll-like receptor expression within the tumor microenvironment. Front Oncol 2024; 14:1308651. [PMID: 38476365 PMCID: PMC10928891 DOI: 10.3389/fonc.2024.1308651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/30/2024] [Indexed: 03/14/2024] Open
Abstract
Toll-like receptors (TLRs) are well-known for their role in cancer development as well as in directing anti-tumor immunity. Because TLRs have also been implicated in the innate recognition of the influenza virus, it was of great interest to investigate the potential TLRs' contribution to the reduction in tumor growth following intratumoral injection of an unadjuvanted influenza vaccine and the lack of antitumor response from an adjuvanted vaccine. In our previous publication, we showed that the unadjuvanted flu vaccine modulates TLR7 expression leading to anti-tumor response in a murine model of melanoma. Here, we show that the unadjuvanted and adjuvanted flu vaccines robustly stimulate different sets of TLRs, TLR3 and TLR7, and TLR4 and TLR9, respectively. In addition, the reduction in tumor growth and improved survival from intratumoral administration of the unadjuvanted vaccine was found to be diminished in TLR7-deficient mice. Finally, we observed that both vaccines have the capacity to modulate TLR expression on both innate and adaptive immune cells. Our findings add to the mechanistic understanding of the parameters that influence tumor outcomes in unadjuvanted and adjuvanted influenza vaccines.
Collapse
Affiliation(s)
- Kajal H. Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL, United States
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL, United States
| | - Eileena F. Giurini
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL, United States
| | - Andrew Zloza
- Division of Surgical Oncology, Department of Surgery, The University of Texas Medical Branch, Galveston, TX, United States
- Division of Translational and Precision Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
10
|
Bertoncini-Silva C, Fassini PG, Carlos D, de Paula NA, Ramalho LNZ, Rodrigues Giuliani M, Pereira ÍS, Guimarães JB, Suen VMM. The Dose-Dependent Effect of Curcumin Supplementation on Inflammatory Response and Gut Microbiota Profile in High-Fat Fed C57BL/6 Mice. Mol Nutr Food Res 2023; 67:e2300378. [PMID: 37818762 DOI: 10.1002/mnfr.202300378] [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: 06/07/2023] [Revised: 09/01/2023] [Indexed: 10/13/2023]
Abstract
SCOPE The prevalence of obesity has increased, with excessive consumption of high-fat foods being one of the primary causes. Curcumin, a polyphenol extracted from Curcuma longa L., exhibits anti-inflammatory activity. The study aims to investigate the effects of curcumin supplementation in different doses on the biochemical profile, inflammatory response, and gut microbiota profile in mice that are fed with high-fat diet (HFD). METHODS AND RESULTS C57BL/6 male mice are fed a standard diet, or a HFD with or without different doses of curcumin (50, 250, and 500 mg kg-1 of body weight). Throughout the experimental period, food intake and body weight are assessed weekly. At euthanasia, blood, stool, and tissue samples are collected for biochemical, histological, and molecular analyses. Curcumin increases the IL-10 protein expression in the white adipose tissue. In the liver, there is a reduction in tumor necrosis factor alpha (TNF-α) and an increase in IL-10 gene expression. Also, curcumin promotes the growth of butyrogenic bacteria, such as Clostridium clusters IV and XIVa. CONCLUSIONS The findings suggest that curcumin has the potential to improve the inflammatory response and modulate healthy gut microbiota. Further studies are needed to clarify the role of curcumin as a preventive and effective strategy for obesity.
Collapse
Affiliation(s)
- Caroline Bertoncini-Silva
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Priscila Giacomo Fassini
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Daniela Carlos
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Natália Aparecida de Paula
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Leandra Náira Zambelli Ramalho
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Marina Rodrigues Giuliani
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Ítalo Sousa Pereira
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Jhefferson Barbosa Guimarães
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Vivian Marques Miguel Suen
- Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900 Monte Alegre, CEP: 14049-900, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
11
|
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
|
12
|
Colleselli K, Stierschneider A, Wiesner C. An Update on Toll-like Receptor 2, Its Function and Dimerization in Pro- and Anti-Inflammatory Processes. Int J Mol Sci 2023; 24:12464. [PMID: 37569837 PMCID: PMC10419760 DOI: 10.3390/ijms241512464] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
While a certain level of inflammation is critical for humans to survive infection and injury, a prolonged inflammatory response can have fatal consequences. Pattern recognition Toll-like receptors (TLRs) are key players in the initiation of an inflammatory process. TLR2 is one of the most studied pattern recognition receptors (PRRs) and is known to form heterodimers with either TLR1, TLR4, TLR6, and TLR10, allowing it to recognize a wide range of pathogens. Although a large number of studies have been conducted over the past decades, there are still many unanswered questions regarding TLR2 mechanisms in health and disease. In this review, we provide an up-to-date overview of TLR2, including its homo- and heterodimers. Furthermore, we will discuss the pro- and anti-inflammatory properties of TLR2 and recent findings in prominent TLR2-associated infectious and neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Christoph Wiesner
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria
| |
Collapse
|
13
|
Ait Djebbara S, Mcheik S, Percier P, Segueni N, Poncelet A, Truyens C. The macrophage infectivity potentiator of Trypanosoma cruzi induces innate IFN-γ and TNF-α production by human neonatal and adult blood cells through TLR2/1 and TLR4. Front Immunol 2023; 14:1180900. [PMID: 37304288 PMCID: PMC10250606 DOI: 10.3389/fimmu.2023.1180900] [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: 03/06/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
We previously identified the recombinant (r) macrophage (M) infectivity (I) potentiator (P) of the protozoan parasite Trypanosoma cruzi (Tc) (rTcMIP) as an immuno-stimulatory protein that induces the release of IFN-γ, CCL2 and CCL3 by human cord blood cells. These cytokines and chemokines are important to direct a type 1 adaptive immune response. rTcMIP also increased the Ab response and favored the production of the Th1-related isotype IgG2a in mouse models of neonatal vaccination, indicating that rTcMIP could be used as a vaccine adjuvant to enhance T and B cell responses. In the present study, we used cord and adult blood cells, and isolated NK cells and human monocytes to investigate the pathways and to decipher the mechanism of action of the recombinant rTcMIP. We found that rTcMIP engaged TLR1/2 and TLR4 independently of CD14 and activated the MyD88, but not the TRIF, pathway to induce IFN-γ production by IL-15-primed NK cells, and TNF-α secretion by monocytes and myeloid dendritic cells. Our results also indicated that TNF-α boosted IFN-γ expression. Though cord blood cells displayed lower responses than adult cells, our results allow to consider rTcMIP as a potential pro-type 1 adjuvant that might be associated to vaccines administered in early life or later.
Collapse
Affiliation(s)
- Sarra Ait Djebbara
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Saria Mcheik
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pauline Percier
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Service Immune Response, Sciensano, Brussels, Belgium
| | - Noria Segueni
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Antoine Poncelet
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
14
|
Rudyk M, Hurmach Y, Serhiichuk T, Akulenko I, Skivka L, Berehova T, Ostapchenko L. Multi-probiotic consumption sex-dependently interferes with MSG-induced obesity and concomitant phagocyte pro-inflammatory polarization in rats: Food for thought about personalized nutrition. Heliyon 2023; 9:e13381. [PMID: 36816299 PMCID: PMC9932736 DOI: 10.1016/j.heliyon.2023.e13381] [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: 06/23/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Epidemic scope which obesity has reached in many countries necessitates shifting the emphasis in medicine from traditional reaction to individualized and personalized prevention. Numerous trials convincingly prove sexual dimorphism of obesity in morbidity, pathophysiology, comorbidity, outcomes and prophylaxis efficacy. Obesity is characterized by chronic systemic low-grade inflammation that creates the preconditions for the emergence of numerous comorbidities. Leading role in the initiation, propagation and resolution of inflammation belongs to tissue resident and circulating phagocytes. The outcome of inflammation largely depends on phagocyte functional polarization, which in turn is governed by environmental stimuli. Gut microbiota (GM), whose disturbances are one of the key pathogenetic features in obesity, substantially affect phagocyte functions and can either aggravate or calm obesity-associated inflammation. Probiotics possess promising physiological functions, including microbiota-restoring and anti-inflammatory traits, that may possibly help prevent obesity. However, sex-specific effects of probiotic supplementation for targeted obesity prevention remain unknown. The aim of the current study was aimed to compare the effect of multi-probiotic preparation used in prophylactic regimen on the adiposity, profile of culturable GM and its short-chain fatty acids as well as on functional profile of phagocytes from different locations in male and female rats with monosodium glutamate (MSG)-induced obesity. Obesity was induced by neonatal MSG injections in male and female rats, who were given the multi-species probiotic during juvenile and adult developmental stages. Culturable fecal and mucosa-associated microbiota of the intestine were examined using selective diagnostic media. Short-chain fatty acid profile in fecal samples was determined by GC-MS. Phagocyte functional profile was evaluated using flow cytometry and colorimetric methods. Probiotic supplementation after the administration of MSG prevented weight gain and fat accumulation, inflammatory phagocyte activation and alterations in GM in female rats. In male MSG-injected rats, probiotic supplementation restricted but did not prevent weight gain and fat deposition, alleviated but did not prevent systemic inflammation, prevented the alterations in GM, but with residual imbalance in the ratio of obligate anaerobic to facultative anaerobic bacteria. Our findings emphasize the necessity of sex-centered approaches to the prophylactic use of probiotics in obesity in the context of predictive preventive and personalized medicine.
Collapse
Affiliation(s)
- Mariia Rudyk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine,Corresponding author.
| | - Yevheniia Hurmach
- Bogomolets National Medical University, 13, T. Shevchenko Blvd, Kyiv, 01601, Ukraine
| | - Tetiana Serhiichuk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Iryna Akulenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Tetiana Berehova
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Liudmyla Ostapchenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| |
Collapse
|
15
|
Bearoff F, Dhavale D, Kotzbauer P, Kortagere S. Aggregated alpha-synuclein transcriptionally activates pro-inflammatory canonical and non-canonical NF-κB signaling pathways in peripheral monocytic cells. Mol Immunol 2023; 154:1-10. [PMID: 36571978 PMCID: PMC9905308 DOI: 10.1016/j.molimm.2022.12.006] [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: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by chronic neuroinflammation, loss of dopaminergic neurons in the substantia nigra, and in several cases accumulation of alpha-synuclein fibril (α-syn) containing Lewy-bodies (LBs). Peripheral inflammation may play a causal role in inducing and perpetuating neuroinflammation in PD and accumulation of fibrillar α-syn has been reported at several peripheral sites including the gut and liver. Peripheral fibrillar α-syn may induce activation of monocytes via recognition by toll-like receptors (TLRs) and stimulation of downstream NF-κB signaling; however, the specific mechanism by which this occurs is not defined. In this study we utilized the THP-1 monocytic cell line to model the peripheral transcriptional response to preformed fibrillar (PFF) α-syn. Compared to monomeric α-syn, PFF α-syn displays overt inflammatory gene upregulation and pathway activation including broad pan-TLR signaling pathway activation and increases in TNF and IL1B gene expression. Notably, the non-canonical NF-κB signaling pathway gene and PD genome wide association study (GWAS) candidate NFKB2 was upregulated. Additionally, non-canonical NF-κB activation-associated RANK and CD40 pathways were also upregulated. Transcriptional-phenotype analysis suggests PFFs induce transcriptional programs associated with differentiation of monocytes towards macrophages and osteoclasts via non-canonical NF-κB signaling as a potential mechanism in which myeloid/monocyte cells may contribute to peripheral inflammation and pathogenesis in PD.
Collapse
Affiliation(s)
- Frank Bearoff
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Dhruva Dhavale
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Paul Kotzbauer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, United States.
| |
Collapse
|
16
|
de Campos GY, Oliveira-Brito PKM, Guimarães JG, da Costa LS, Lazo Chica JE, da Silva TA. Adjuvant Pam3CSk4 does not improve the immunization against Cryptococcus gattii infection in C57BL/6 mice. PeerJ 2023; 11:e14778. [PMID: 36743957 PMCID: PMC9897066 DOI: 10.7717/peerj.14778] [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: 08/08/2022] [Accepted: 01/03/2023] [Indexed: 02/01/2023] Open
Abstract
Background Cryptococcosis is a relevant invasive fungal infection that affects immunocompromised and immunocompetent individuals when caused by Cryptococcus gattii. Host innate and adaptive immune responses can be subverted by C. gattii, that blocks the differentiation of T helper (Th) 1 and Th17 cells, which are involved in the protection against cryptococcosis. Moreover, the macrophage polarization is modulated by C. gattii infection that requires a balance in the macrophage subsets to control the C. gattii infection. Toll-like receptor (TLR) 2 agonists are important immunomodulators favoring a pro-inflammatory response with potential fungicidal activity, and TLR2 agonists have been used as adjuvants in vaccines against infections caused by bacteria or viruses. Therefore, this work aimed to evaluate the immunomodulatory effect of the tripalmitoyl lipopeptide S-glycerol cysteine (Pam3CSK4 or P3C4), a TLR2 agonist, as an adjuvant in the vaccination against C. gattii infection. Methods and Results C57BL/6 mice were immunized with 2 × 107 inactivated yeasts of C. gattii via intranasal route on day 1, 14 and 28 (Immunized group). Immunization was associated with 1µg or 10µg of adjuvant P3C4 (Immunized+P3C4-1µg or Immunized+P3C4-10 µg), followed by C. gattii infection on day 42 after the immunization protocol. Immunized+P3C4-1 µg group had reduced levels of IgG1, IgG2a and IgA and no significant difference in the IgG and IgM anti-GXM antibody titer was detected, compared to the Immunized group. High levels of IL-17 and IL-1β in lung tissue of mice from the Immunized+P3C4-1µg group did not promote a predominance of Th17 cells, in contrast, the frequency of TLR2+ cells was increased in immunized mice that received 1 µg of P3C4. The reduction in the relative expression of T-bet and high levels of Foxp3 detected in the lungs of the Immunized+P3C4-1µg group suggest a prevalence of regulatory T cells in the tissue, which did not contribute to the control of C. gattii infection. The immunization protocol associated with 10 µg of adjuvant P3C4 induced high levels of IL-17 in the lung tissue, whereas the levels of pro-inflammatory cytokines were downregulated. To evaluate the effect of adjuvant P3C4 in the control of C. gattii infection, quantification of the fungal burden in the lungs was performed by the CFU assay, and the groups with adjuvant P3C4 showed a pulmonary C. gattii burden that was not significantly altered when compared with the immunized group. The mice that received 1 µg of adjuvant P3C4 had a lower percentage of inflammatory infiltrate in the lungs. Conclusion The immunomodulatory effect of P3C4, associated with the immunization protocol, plays an imbalance between pro- and anti-inflammatory response in the lungs that did not favor a protection against C. gattii infection, which is related to the immune response characterized by a suppressive/regulatory profile in the pulmonary microenvironment after C. gattii infection.
Collapse
Affiliation(s)
- Gabriela Yamazaki de Campos
- Department of Cell and Molecular Biology and Pathogenic Bioagents, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Júlia Garcia Guimarães
- Department of Cell and Molecular Biology and Pathogenic Bioagents, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Letícia Serafim da Costa
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Javier Emílio Lazo Chica
- Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Thiago Aparecido da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
17
|
Barboza BR, Thomaz SMDO, Junior ADC, Espreafico EM, Miyamoto JG, Tashima AK, Camacho MF, Zelanis A, Roque-Barreira MC, da Silva TA. ArtinM Cytotoxicity in B Cells Derived from Non-Hodgkin's Lymphoma Depends on Syk and Src Family Kinases. Int J Mol Sci 2023; 24:ijms24021075. [PMID: 36674590 PMCID: PMC9863955 DOI: 10.3390/ijms24021075] [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: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023] Open
Abstract
Receptors on the immune cell surface have a variety of glycans that may account for the immunomodulation induced by lectins, which have a carbohydrate recognition domain (CRD) that binds to monosaccharides or oligosaccharides in a specific manner. ArtinM, a D-mannose-binding lectin obtained from Artocarpus heterophyllus, has affinity for the N-glycans core. Immunomodulation by ArtinM toward the Th1 phenotype occurs via its interaction with TLR2/CD14 N-glycans on antigen-presenting cells, as well as recognition of CD3γ N-glycans on murine CD4+ and CD8+ T cells. ArtinM exerts a cytotoxic effect on Jurkat human leukemic T-cell line and human myeloid leukemia cell line (NB4). The current study evaluated the effects of ArtinM on murine and human B cells derived from non-Hodgkin’s lymphoma. We found that murine B cells are recognized by ArtinM via the CRD, and the ArtinM stimulus did not augment the proliferation rate or production of IL-2. However, murine B cell incubation with ArtinM augmented the rate of apoptosis, and this cytotoxic effect of ArtinM was also seen in human B cell-lines sourced from non-Hodgkin’s lymphoma Raji cell line. This cytotoxic effect was inhibited by the phosphatase activity of CD45 on Lck, and the protein kinases of the Src family contribute to cell death triggered by ArtinM.
Collapse
Affiliation(s)
- Bruno Rafael Barboza
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Sandra Maria de Oliveira Thomaz
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Airton de Carvalho Junior
- Laboratory of Cell and Molecular Biology of Cancer, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Enilza Maria Espreafico
- Laboratory of Cell and Molecular Biology of Cancer, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Jackson Gabriel Miyamoto
- Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), Sao Paulo 04021-001, SP, Brazil
| | - Alexandre Keiji Tashima
- Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), Sao Paulo 04021-001, SP, Brazil
| | - Maurício Frota Camacho
- Functional Proteomics Laboratory, Department of Science and Technology, Federal University of São Paulo (ICT-UNIFESP), São José dos Campos 04021-001, SP, Brazil
| | - André Zelanis
- Functional Proteomics Laboratory, Department of Science and Technology, Federal University of São Paulo (ICT-UNIFESP), São José dos Campos 04021-001, SP, Brazil
| | - Maria Cristina Roque-Barreira
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Thiago Aparecido da Silva
- Laboratory of Immunotherapy of Invasive Fungal Infections, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
- Correspondence: or ; Tel.: +55-16-3315-3049
| |
Collapse
|
18
|
Microglia-Neutrophil Interactions Drive Dry AMD-like Pathology in a Mouse Model. Cells 2022; 11:cells11223535. [PMID: 36428965 PMCID: PMC9688699 DOI: 10.3390/cells11223535] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/18/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
In dry age-related macular degeneration (AMD), inflammation plays a key role in disease pathogenesis. Innate immune cells such as microglia and neutrophils infiltrate the sub-retinal space (SRS) to induce chronic inflammation and AMD progression. But a major gap in our understanding is how these cells interact with each other in AMD. Here, we report a novel concept of how dynamic interactions between microglia and neutrophils contribute to AMD pathology. Using well-characterized genetically engineered mouse models as tools, we show that in the diseased state, retinal pigmented epithelial (RPE) cells trigger pro-inflammatory (M1) transition in microglia with diminished expression of the homeostatic marker, CX3CR1. Activated microglia localize to the SRS and regulate local neutrophil function, triggering their activation and thereby inducing early RPE changes. Ligand receptor (LR)-loop analysis and cell culture studies revealed that M1 microglia also induce the expression of neutrophil adhesion mediators (integrin β1/α4) through their interaction with CD14 on microglia. Furthermore, microglia-induced neutrophil activation and subsequent neutrophil-mediated RPE alterations were mitigated by inhibiting Akt2 in microglia. These results suggest that the Akt2 pathway in microglia drives M1 microglia-mediated neutrophil activation, thereby triggering early RPE degeneration and is a novel therapeutic target for early AMD, a stage without treatment options.
Collapse
|
19
|
Expression of Antimicrobic Peptide Piscidin1 in Gills Mast Cells of Giant Mudskipper Periophthalmodon schlosseri (Pallas, 1770). Int J Mol Sci 2022; 23:ijms232213707. [PMID: 36430187 PMCID: PMC9692400 DOI: 10.3390/ijms232213707] [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: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
The amphibious teleost Giant mudskipper (Periophthalmodon schlosseri, Pallas 1770) inhabit muddy plains and Asian mangrove forests. It spends more than 90% of its life outside of the water, using its skin, gills, and buccal-pharyngeal cavity mucosa to breathe in oxygen from the surrounding air. All vertebrates have been found to have mast cells (MCs), which are part of the innate immune system. These cells are mostly found in the mucous membranes of the organs that come in contact with the outside environment. According to their morphology, MCs have distinctive cytoplasmic granules that are released during the degranulation process. Additionally, these cells have antimicrobial peptides (AMPs) that fight a variety of infections. Piscidins, hepcidins, defensins, cathelicidins, and histonic peptides are examples of fish AMPs. Confocal microscopy was used in this study to assess Piscidin1 expression in Giant Mudskipper branchial MCs. Our results demonstrated the presence of MCs in the gills is highly positive for Piscidin1. Additionally, colocalized MCs labeled with TLR2/5-HT and Piscidin1/5-HT supported our data. The expression of Piscidin1 in giant mudskipper MCs highlights the involvement of this peptide in the orchestration of teleost immunity, advancing the knowledge of the defense system of this fish.
Collapse
|
20
|
Wöhnke E, Cackett G, Werner F, Blome S, Mettenleiter TC, Karger A. Proteome Analysis of Swine Macrophages after Infection with Two Genotype II African Swine Fever Isolates of Different Pathogenicity. Viruses 2022; 14:v14102140. [PMID: 36298696 PMCID: PMC9607119 DOI: 10.3390/v14102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Since the introduction of a highly pathogenic genotype II isolate of the African swine fever virus (ASFV) into Georgia in 2007, African swine fever (ASF) has gone panzootic. Outbreaks have been reported in Europe, Asia and, more recently, Latin America. Thus, ASFV has become a major threat to the pig industry worldwide, as broadly applicable vaccines are not available. While the majority of ASFV strains show high virulence in domestic pigs and wild boar, variations within the ASFV genome have resulted in the emergence of attenuated strains with low or moderate virulence. However, the molecular basis of the differences in virulence has not yet been discovered. To reveal virulence-associated protein expression patterns, we analysed the proteomes of the natural target cells of ASFV, primary porcine macrophages, after infection with two genotype II ASFV strains displaying high (Armenia 2008) and moderate (Estonia 2014) virulence using quantitative mass spectrometry. Very similar expression patterns were observed for the viral genes, and any differences were limited to the deletions within the Estonia 2014 genome. In addition to the canonical ASFV proteins, twelve novel protein products from recently described transcripts were confirmed in both isolates. Pathway analyses showed that both isolates evoked a similar host proteome response, despite their difference in virulence. However, subtle differences in the manipulation of the proteins involved in the proinflammatory response mediated by the MAPK14/p38 signalling cascade were observed.
Collapse
Affiliation(s)
- Elisabeth Wöhnke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
| | - Gwenny Cackett
- Institute for Structural and Molecular Biology, Darwin Building, University College London, Gower Street, London WC1E 6BT, UK
| | - Finn Werner
- Institute for Structural and Molecular Biology, Darwin Building, University College London, Gower Street, London WC1E 6BT, UK
| | - Sandra Blome
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
| | - Thomas C. Mettenleiter
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
| | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald, Germany
- Correspondence: ; Tel.: +49-38351-7-1247
| |
Collapse
|
21
|
Aripova N, Duryee MJ, Hunter CD, Ryan EM, Daubach EC, Jones SQ, Bierman MM, Ragland AS, Mitra A, England BR, Romberger DJ, Thiele GM, Mikuls TR. Peptidyl arginine deiminase expression and macrophage polarization following stimulation with citrullinated and malondialdehyde-acetaldehyde modified fibrinogen. Int Immunopharmacol 2022; 110:109010. [PMID: 35785731 DOI: 10.1016/j.intimp.2022.109010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Post-translational modifications of extracellular matrix proteins such as fibrinogen may lead to tolerance loss and have been implicated in rheumatoid arthritis (RA) pathogenesis. The purpose of this study was to determine whether fibrinogen (FIB) modified with citrulline (CIT), malondialdehyde-acetaldehyde (MAA) or both leads to altered macrophage polarization, peptidyl arginine deiminase (PAD) expression, or production of citrullinated proteins. METHODS PMA-treated U-937 cells (M0 cells) were stimulated with MAA, CIT or MAA-CIT modified FIB. Macrophage (M1/M2) phenotypes were evaluated by flow cytometry, RT-PCR, and ELISA. PAD enzyme expression and protein citrullination was evaluated using RT-PCR and Western Blot. RESULTS Flow cytometry revealed that M0 macrophages stimulated with FIB-MAA-CIT resulted in mixed M1/M2 phenotypes as demonstrated by cell surface expression and mRNA levels of CD14, CD192, CD163, and CD206 (p < 0.001 vs. others), and the release of IL-18, IP-10, CCL22, and IL-13 (p < 0.001 vs. others). While FIB-MAA treated M0 cells demonstrated a mixed M1/M2 phenotype, cytokine and cell surface markers differed from FIB-MAA-CIT. Finally, M0 cells treated with FIB-CIT demonstrated markers and cytokines consistent with only the M1-like phenotype. Exposure of M0 cells to FIB-MAA-CIT (at 48 h) and FIB-MAA (at 24 h) led to increased mRNA expression and protein expression of PAD2 (p < 0.001) with increased protein citrullination. CONCLUSION These findings suggest that MAA-modification and citrullination of FIB, in isolation or combination, yield specific effects on macrophage polarization, PAD expression and citrullination that ultimately may induce inflammatory and fibrotic responses associated with RA.
Collapse
Affiliation(s)
- Nozima Aripova
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael J Duryee
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Carlos D Hunter
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Evan M Ryan
- Department of Otolaryngology - Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric C Daubach
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Spencer Q Jones
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Madison M Bierman
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Austin S Ragland
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ananya Mitra
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bryant R England
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Debra J Romberger
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Ted R Mikuls
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA; Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
22
|
Lee HR, Lee S, Yoo IS, Yoo SJ, Kwon MH, Joung CI, Park JA, Wook Kang S, Kim J. CD14+ monocytes and soluble CD14 of synovial fluid are associated with osteoarthritis progression. Arch Rheumatol 2022; 37:335-343. [PMID: 36589618 PMCID: PMC9791551 DOI: 10.46497/archrheumatol.2022.9078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/02/2021] [Indexed: 01/15/2023] Open
Abstract
Objectives This study aims to investigate the role of cluster of differentiation 14 (CD14) expressed monocytes and soluble CD14-mediated pathway in the synovial inflammation of knee osteoarthritis (OA). Patients and methods Between May 2012 and July 2013, a total of 35 patients with knee OA (9 males, 26 females; mean age: 66.3±8.8 years; range, 52 to 79 years) were included in this cross-sectional study. Synovial fluid was obtained from knee joints of 35 OA patients. The CD14+ monocytes from synovial fluid mononuclear cells (SFMCs) were isolated using the MACS. The fibroblast-like synoviocytes (FLSs) isolated from knee joint tissue were incubated with recombinant CD14 and lipopolysaccharide (LPS) for 24 h. Cytokine profiling was performed with the Luminex® Performance Assay or magnetic bead panel kit. The expression of CD14 and CD16 was analyzed by immunohistochemistry and flow cytometry. Results The concentration of sCD14 in synovial fluid was correlated with the interleukin-6 (IL-6) level (n=35) (ρ=0.654, p<0.001). The culture supernatants of CD14+ monocytes isolated from SFMC (n=15) showed a correlation between sCD14 and IL-6 (ρ=0.784, p=0.001), along with complement component 3 (ρ=0.756, p=0.010), IL-1b (ρ=0.652, p=0.012), and tumor necrosis factor-alpha (ρ=0.806, p=0.001). Following recombinant CD14 and LPS treatment, OA FLS synergistically enhanced the secretion of IL-6, IL-8, and matrix metalloproteinase 3 (n=3, p<0.05). In five paired-samples from identical patients, the proportions of CD14+ monocytes were significantly elevated in recurred synovial fluid compared to those in initial synovial fluid (p=0.043). When monocyte subsets were analyzed in SFMC (n=26), CD14+CD16+monocytes were abundant (p=0.019) and had higher toll-like receptor 4 expression than CD14+CD16- (p<0.001). Conclusion Our study results suggest that CD14+ monocytes and the sCD14-mediated pathway play an important role in OA aggravation through inflammatory cytokine secretion.
Collapse
Affiliation(s)
- Ha-Reum Lee
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
,
Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sunyoung Lee
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - In Seol Yoo
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Su-Jin Yoo
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Mi-Hye Kwon
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Republic of Korea
| | - Chung-il Joung
- Department of Internal Medicine, Konyang University School of Medicine, Daejeon, Republic of Korea
| | - Ji Ah Park
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
,
Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Jinhyun Kim
- Department of Internal Medicine, Division of Rheumatology, Chungnam National University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Tobita K, Meguro R. Bacillus subtilis BN strain promotes Th1 response via Toll-like receptor 2 in polarized mouse M1 macrophage. J Food Biochem 2022; 46:e14046. [PMID: 34997586 DOI: 10.1111/jfbc.14046] [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: 07/24/2021] [Revised: 11/07/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022]
Abstract
Bacillus subtilis BN strain (BN strain) was isolated from natto, a traditional Japanese fermented soybean food product. The present study investigated the Th1 responses of the BN strain on a mouse macrophage cell line, J774.1. In cell cultures, the BN strain (spore cell cultured in Schaeffer's sporulation media) significantly increased the production of interleukin (IL-)12 protein. The BN strain induced the mRNA expression of M1 polarization genes, such as inducible nitric oxide synthase and IL-12p40 mRNA, and suppressed the mRNA expression of intracellular marker genes of M2 polarization, such as arginase 1 mRNA. The BN strain downregulated the mRNA expression of Toll-like receptor 4 (TLR4), while it upregulated the mRNA expression of TLR2, MyD88, and nuclear factor kappa B (NF-κB). The production of IL-12 protein induced by the BN strain was decreased by inhibitors of MyD88, NF-κB, and IκB kinase. Moreover, the production of IL-12 was strongly suppressed by neutralizing antibody against TLR2. These results suggest that the BN strain promotes Th1 response via TLR2 signal in mouse M1 macrophage. PRACTICAL APPLICATIONS: Bacillus subtilis is known to have beneficial effects for the host. B. subtilis BN stain (BN strain) was isolated from natto, a traditional Japanese fermented soybean food product. The effects of the BN strain on the Th1 response in macrophage cell cultures were investigated in this work. We found that the spore cells of BN strain promoted the production of Th1-type cytokine, and induced macrophage M1 polarization via Toll-like receptor 2. This study can serve as a significant reference for the development of functional food and feed with immunostimulatory effects. Over time, new food and feed products containing the BN strain may emerge, such as Juice, powder, and tablet.
Collapse
Affiliation(s)
- Keisuke Tobita
- Industrial Technology Innovation Center of Ibaraki Prefecture, Nagaoka, Japan
| | | |
Collapse
|
24
|
Getachew A, Hussain M, Huang X, Li Y. Toll-like receptor 2 signaling in liver pathophysiology. Life Sci 2021; 284:119941. [PMID: 34508761 DOI: 10.1016/j.lfs.2021.119941] [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: 05/08/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022]
Abstract
Chronic liver diseases (CLD) are among the major cause of mortality and morbidity worldwide. Despite current achievements in the area of hepatitis virus, chronic alcohol abuse and high-fat diet are still fueling an epidemic of severe liver disease, for which, an effective therapy has yet not been discovered. In particular, the therapeutic regimens that could prevent the progression of fibrosis and, in turn, aid cirrhotic liver to develop a robust regenerative capability are intensively needed. To this context, a better understanding of the signaling pathways regulating hepatic disease development may be of critical value. In general, the liver responds to various insults with an orchestrated healing process involving variety of signaling pathways. One such pathway is the TLR2 signaling pathway, which essentially regulates adult liver pathogenesis and thus has emerged as an attractive target to treat liver disease. TLR2 is expressed by different liver cells, including Kupffer cells (KCs), hepatocytes, and hepatic stellate cells (HSCs). From a pathologic perspective, the crosstalk between antigens and TLR2 may preferentially trigger a distinctive set of signaling mechanisms in these liver cells and, thereby, induce the production of inflammatory and fibrogenic cytokines that can initiate and prolong liver inflammation, ultimately leading to fibrosis. In this review, we summarize the currently available evidence regarding the role of TLR2 signaling in hepatic disease progression. We first elaborate its pathological involvement in liver-disease states, such as inflammation, fibrosis, and cirrhosis. We then discuss how therapeutic targeting of this pathway may help to alleviate its disease-related functioning.
Collapse
Affiliation(s)
- Anteneh Getachew
- Institute of Public Health, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Muzammal Hussain
- Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xinping Huang
- Institute of Public Health, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yinxiong Li
- Institute of Public Health, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China.
| |
Collapse
|
25
|
Huang R, Hu Z, Chen X, Cao Y, Li H, Zhang H, Li Y, Liang L, Feng Y, Wang Y, Su W, Kong Z, Melgiri ND, Jiang L, Li X, Du J, Chen Y. The Transcription Factor SUB1 Is a Master Regulator of the Macrophage TLR Response in Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004162. [PMID: 34378353 PMCID: PMC8498911 DOI: 10.1002/advs.202004162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/24/2021] [Indexed: 12/26/2022]
Abstract
Toll-like receptor 2 and 4 (TLR2, TLR4) signaling is implicated in atherosclerotic plaque formation. The two-stage master regulator Virtual Inference of Protein-activity by Enriched Regulon (VIPER) analysis of macrophage TLR2 and TLR4 signature genes integrated with coexpression network genes derived from 371 patient-derived carotid specimens identifies activated RNA polymerase II transcriptional coactivator p15 (SUB1/Sub1, PC4) as a master regulon in the atherogenic TLR response. It is found that TLR2 and TLR4 signaling is proinflammatory and proatherosclerotic in chow-fed apolipoprotein E-deficient (ApoE-/- ) mice. Through transgenic myeloid-specific Sub1 knockout in ApoE-/- mice, it is discovered that these proatherosclerotic effects of TLR2 and TLR4 signaling are mediated by Sub1. Sub1 knockout in macrophages enhances anti-inflammatory M2 macrophage polarization and cholesterol efflux. Irradiated low density lipoprotein receptor-deficient (Ldlr-/- ) mice transplanted with Sub1-/- murine bone marrow display reduced atherosclerosis. Promoter analysis reveals Sub1-dependent activation of interferon regulatory factor 1 (Irf1) transcription in a casein kinase 2 (Ck2)-dependent manner, and Sub1-knockout macrophages display decreased Irf1 expression. Artificial Irf1 overexpression in Sub1-knockout macrophages enhances proinflammatory M1 skewing and lowers cholesterol clearance. In conclusion, the TLR master regulon Sub1, and its downstream effect on the transcription factor Irf1, promotes a proinflammatory M1 macrophage phenotype and enhances atherosclerotic burden in vivo.
Collapse
Affiliation(s)
- Rongzhong Huang
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zicheng Hu
- Institute of Ultrasound ImagingThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaorui Chen
- Department of Pulmonary and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Yu Cao
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Hongrong Li
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Hong Zhang
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Yongyong Li
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Liwen Liang
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Yuxing Feng
- Department of Rehabilitation and Pain MedicineThe Ninth People's Hospital of ChongqingChongqing400700China
| | - Ying Wang
- Department of Rehabilitation MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Wenhua Su
- Department of CardiologyThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Zerui Kong
- Department of Cardiothoracic SurgeryThe Affiliated Yan An Hospital of Kunming Medical UniversityKunming650000China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunming650500China
| | - ND Melgiri
- Impactys Foundation for Biomedical ResearchSan DiegoCA92121USA
| | - Lihong Jiang
- Department of Cardiothoracic SurgeryThe First People's Hospital of Yunnan ProvinceKunming650032China
| | - Xingsheng Li
- Department of Geriatric MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Jianlin Du
- Department of CardiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Yunqing Chen
- Department of CardiologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| |
Collapse
|
26
|
Siefert JC, Cioni B, Muraro MJ, Alshalalfa M, Vivié J, van der Poel HG, Schoots IG, Bekers E, Feng FY, Wessels LFA, Zwart W, Bergman AM. The Prognostic Potential of Human Prostate Cancer-Associated Macrophage Subtypes as Revealed by Single-Cell Transcriptomics. Mol Cancer Res 2021; 19:1778-1791. [PMID: 34131070 PMCID: PMC9398107 DOI: 10.1158/1541-7786.mcr-20-0740] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/18/2020] [Accepted: 06/07/2021] [Indexed: 01/07/2023]
Abstract
Macrophages in the tumor microenvironment are causally linked with prostate cancer development and progression, yet little is known about their composition in neoplastic human tissue. By performing single cell transcriptomic analysis of human prostate cancer resident macrophages, three distinct populations were identified in the diseased prostate. Unexpectedly, no differences were observed between macrophages isolated from the tumorous and nontumorous portions of the prostatectomy specimens. Markers associated with canonical M1 and M2 macrophage phenotypes were identifiable, however these were not the main factors defining unique subtypes. The genes selectively associated with each macrophage cluster were used to develop a gene signature which was highly associated with both recurrence-free and metastasis-free survival. These results highlight the relevance of tissue-specific macrophage subtypes in the tumor microenvironment for prostate cancer progression and demonstrates the utility of profiling single-cell transcriptomics in human tumor samples as a strategy to design gene classifiers for patient prognostication. IMPLICATIONS: The specific macrophage subtypes present in a diseased human prostate have prognostic value, suggesting that the relative proportions of these populations are related to patient outcome. Understanding the relative contributions of these subtypes will not only inform patient prognostication, but will enable personalized immunotherapeutic strategies to increase beneficial populations or reduce detrimental populations.
Collapse
Affiliation(s)
- Joseph C Siefert
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Bianca Cioni
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mauro J Muraro
- Single Cell Discoveries B.V., the Netherlands.,Hubrecht Institute-KNAW and University Medical Center Utrecht, the Netherlands
| | - Mohammed Alshalalfa
- Department of Radiation Oncology, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Judith Vivié
- Single Cell Discoveries B.V., the Netherlands.,Hubrecht Institute-KNAW and University Medical Center Utrecht, the Netherlands
| | - Henk G van der Poel
- Division of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ivo G Schoots
- Department of Radiology and Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Elise Bekers
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Felix Y Feng
- Department of Radiation Oncology, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands. .,Oncode Institute, the Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, the Netherlands. .,Oncode Institute, the Netherlands
| | - Andries M Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, the Netherlands. .,Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
27
|
Stanojević S, Blagojević V, Ćuruvija I, Vujić V. Lactobacillus rhamnosus Affects Rat Peritoneal Cavity Cell Response to Stimulation with Gut Microbiota: Focus on the Host Innate Immunity. Inflammation 2021; 44:2429-2447. [PMID: 34505975 DOI: 10.1007/s10753-021-01513-z] [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: 04/18/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022]
Abstract
Gut microbiota contribute to shaping the immune repertoire of the host, whereas probiotics may exert beneficial effects by modulating immune responses. Having in mind the differences in both the composition of gut microbiota and the immune response between rats of Albino Oxford (AO) and Dark Agouti (DA) rat strains, we investigated if intraperitoneal (i.p.) injection of live Lactobacillus rhamnosus (LB) may influence peritoneal cavity cell response to in vitro treatments with selected microbiota in the rat strain-dependent manner. Peritoneal cavity cells from AO and DA rats were lavaged two (d2) and seven days (d7) following i.p. injection with LB and tested for NO, urea, and H2O2 release basally, or upon in vitro stimulation with autologous E.coli and Enterococcus spp. Whereas the single i.p. injection of LB nearly depleted resident macrophages and increased the proportion of small inflammatory macrophages and monocytes on d2 in both rat strains, greater proportion of MHCIIhiCD163- and CCR7+ cells and increased NO/diminished H2O2 release in DA compared with AO rats suggest a more intense inflammatory priming by LB in this rat strain. Even though E.coli- and/or Enterococcus spp.-induced rise in H2O2 release in vitro was abrogated by LB in cells from both rat strains, LB prevented microbiota-induced increase in NO/urea ratio only in cells from AO and augmented it in cells from DA rats. Thus, the immunomodulatory properties may not be constant for particular probiotic bacteria, but shaped by innate immunity of the host.
Collapse
Affiliation(s)
- Stanislava Stanojević
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia. .,Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia.
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia
| | - Ivana Ćuruvija
- Immunology Research Centre "Branislav Janković, Institute of Virology, Vaccines and Sera "Torlak, Belgrade, Serbia
| | - Vesna Vujić
- Department of Chemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
28
|
Li WT, Huang XF, Deng C, Zhang BH, Qian K, He M, Sun TL. Olanzapine Induces Inflammation and Immune Response via Activating ER Stress in the Rat Prefrontal Cortex. Curr Med Sci 2021; 41:788-802. [PMID: 34403105 DOI: 10.1007/s11596-021-2401-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Antipsychotics, in particular olanzapine, are first-line medications for schizophrenia. The prefrontal cortex (PFC) is an important region for antipsychotics' therapeutic effects. The PFC inflammatory and immune pathways are associated with schizophrenia pathogenesis. However, the effect of antipsychotics on the inflammatory and immune pathways in the PFC remains unclear. We aimed to examined the time-dependent effect of olanzapine on inflammatory and immune markers in the PFC of rats. Since the inflammatory and immune pathways are related to endoplasmic reticulum (ER) stress, we further investigated whether or not olanzapine-induced inflammation and immune responses were related to ER stress. METHODS Expression of pro-inflammatory markers including IkappaB kinase β (IKKβ), nuclear factor kappa B (NFκB), tumor necrosis factor α (TNF-α), interleukin-6 (IL-6) and IL-1β, and immune-related proteins including inducible nitric oxide synthase (iNOS), toll-like receptor 2 (TLR2) and cluster of differentiation 14 (CD14) were examined by Western blotting. RESULTS Olanzapine treatments for 1, 8 and 36 days significantly activated the inflammatory IKKβ/NFκB signaling, and increased the expression of TNF-α, IL-6, IL-1β and immune-related proteins such as iNOS, TLR4 and CD14. Olanzapine treatment for 1 day, 8 and 36 days also induced ER stress in the PFC. Co-treatment with an ER stress inhibitor, 4-phenylbutyrate, inhibited olanzapine-induced inflammation and the immune response in the PFC. CONCLUSION These results suggested olanzapine exposure could be a factor that induces central inflammation and immunological abnormities in schizophrenia subjects. Olanzapine induces PFC inflammation and immune response, possibly via activating ER stress signaling.
Collapse
Affiliation(s)
- Wen-Ting Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Chao Deng
- Illawarra Health and Medical Research Institute and Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Bao-Hua Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & the Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Kun Qian
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| | - Tao-Lei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
29
|
Clavijo-Cornejo D, Hernández-González O, Gutierrez M. The current role of NLRP3 inflammasome polymorphism in gout susceptibility. Int J Rheum Dis 2021; 24:1257-1265. [PMID: 34390315 DOI: 10.1111/1756-185x.14205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The NLR family pyrin domain containing 3 (NLRP3) signaling pathway has an important role in inflammation mediated by monosodium urate crystals in gout, and the characterization of single nucleotide polymorphisms (SNPs) have helped to recognize disease susceptibility. OBJECTIVE The aim of this review is to provide an overview of the potential role of the inflammasome gene SNPs as a susceptibility factor for gout, discussing the current evidence available. METHODS This review analyzes the relevant literature in the field of inflammasome SNPs and gout published in the last 10 years. The systematic research was performed in 16 articles, including both the SNPs associated and those not associated with gout, with the goal to have a complete overview. RESULTS Sixty-nine SNPs from 10 different genes have been reported in the literature. Of these, 13 SNPs present association with gout susceptibility in different populations, while 56 have been established as not being associated with the disease. CONCLUSIONS This review is a summary of the potential role of inflammasome gene SNPs and their association with gout risk, all of them related with NLRP3 inflammasome signaling, suggesting these polymorphisms are susceptibility candidates and genetic markers for gout. From the 69 SNPs analyzed in the literature, 13 of them have been associated with gout as follows: NLRP3 (rs3806268 and rs10754558), CARD8 (rs2043211), TLR4 (rs2149356), CD14 (rs2569190), IL-1β (rs1143623), P2RX7 (rs2230911, rs1653624, rs7958316 and rs17525809) and PPARGC1B (rs45520937, rs10491360 and rs7712296) in different populations.
Collapse
Affiliation(s)
- Denise Clavijo-Cornejo
- Division of Musculoskeletal and Rheumatic Diseases, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | | | - Marwin Gutierrez
- Division of Musculoskeletal and Rheumatic Diseases, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| |
Collapse
|
30
|
Sefiani A, Geoffroy CG. The Potential Role of Inflammation in Modulating Endogenous Hippocampal Neurogenesis After Spinal Cord Injury. Front Neurosci 2021; 15:682259. [PMID: 34220440 PMCID: PMC8249862 DOI: 10.3389/fnins.2021.682259] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Currently there are approximately 291,000 people suffering from a spinal cord injury (SCI) in the United States. SCI is associated with traumatic changes in mobility and neuralgia, as well as many other long-term chronic health complications, including metabolic disorders, diabetes mellitus, non-alcoholic steatohepatitis, osteoporosis, and elevated inflammatory markers. Due to medical advances, patients with SCI survive much longer than previously. This increase in life expectancy exposes them to novel neurological complications such as memory loss, cognitive decline, depression, and Alzheimer's disease. In fact, these usually age-associated disorders are more prevalent in people living with SCI. A common factor of these disorders is the reduction in hippocampal neurogenesis. Inflammation, which is elevated after SCI, plays a major role in modulating hippocampal neurogenesis. While there is no clear consensus on the mechanism of the decline in hippocampal neurogenesis and cognition after SCI, we will examine in this review how SCI-induced inflammation could modulate hippocampal neurogenesis and provoke age-associated neurological disorders. Thereafter, we will discuss possible therapeutic options which may mitigate the influence of SCI associated complications on hippocampal neurogenesis.
Collapse
|
31
|
Jeljeli M, Riccio LGC, Chouzenoux S, Moresi F, Toullec L, Doridot L, Nicco C, Bourdon M, Marcellin L, Santulli P, Abrão MS, Chapron C, Batteux F. Macrophage Immune Memory Controls Endometriosis in Mice and Humans. Cell Rep 2021; 33:108325. [PMID: 33147452 DOI: 10.1016/j.celrep.2020.108325] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/22/2020] [Accepted: 10/07/2020] [Indexed: 01/08/2023] Open
Abstract
Endometriosis is a frequent, chronic, inflammatory gynecological disease characterized by the presence of ectopic endometrial tissue causing pain and infertility. Macrophages have a central role in lesion establishment and maintenance by driving chronic inflammation and tissue remodeling. Macrophages can be reprogrammed to acquire memory-like characteristics after antigenic challenge to reinforce or inhibit a subsequent immune response, a phenomenon termed "trained immunity." Here, whereas bacille Calmette-Guérin (BCG) training enhances the lesion growth in a mice model of endometriosis, tolerization with repeated low doses of lipopolysaccharide (LPSlow) or adoptive transfer of LPSlow-tolerized macrophages elicits a suppressor effect. LPSlow-tolerized human macrophages mitigate the fibro-inflammatory phenotype of endometriotic cells in an interleukin-10 (IL-10)-dependent manner. A history of severe Gram-negative infection is associated with reduced infertility duration and alleviated symptoms, in contrast to patients with Gram-positive infection history. Thus, the manipulation of innate immune memory may be effective in dampening hyper-inflammatory conditions, opening the way to promising therapeutic approaches.
Collapse
Affiliation(s)
- Mohamed Jeljeli
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Service d'immunologie biologique, 75014 Paris, France
| | - Luiza G C Riccio
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, 01246903 São Paulo, Brasil
| | - Sandrine Chouzenoux
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Fabiana Moresi
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Laurie Toullec
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Ludivine Doridot
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Carole Nicco
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Mathilde Bourdon
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Louis Marcellin
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Pietro Santulli
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Mauricio S Abrão
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, 01246903 São Paulo, Brasil
| | - Charles Chapron
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Frédéric Batteux
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Service d'immunologie biologique, 75014 Paris, France.
| |
Collapse
|
32
|
Park DD, Chen J, Kudelka MR, Jia N, Haller CA, Kosaraju R, Premji AM, Galizzi M, Nairn AV, Moremen KW, Cummings RD, Chaikof EL. Resident and elicited murine macrophages differ in expression of their glycomes and glycan-binding proteins. Cell Chem Biol 2021; 28:567-582.e4. [PMID: 33378651 PMCID: PMC8052306 DOI: 10.1016/j.chembiol.2020.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/17/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022]
Abstract
The pleiotropic functions of macrophages in immune defense, tissue repair, and maintenance of tissue homeostasis are supported by the heterogeneity in macrophage sub-populations that differ both in ontogeny and polarization. Although glycans and glycan-binding proteins (GBPs) are integral to macrophage function and may contribute to macrophage diversity, little is known about the factors governing their expression. Here, we provide a resource for characterizing the N-/O-glycomes of various murine peritoneal macrophage sub-populations, demonstrating that glycosylation primarily reflects developmental origin and, to a lesser degree, cellular polarization. Furthermore, comparative analysis of GBP-coding genes in resident and elicited macrophages indicated that GBP expression is consistent with specialized macrophage functions and correlates with specific types of displayed glycans. An integrated, semi-quantitative approach was used to confirm distinct expression patterns of glycans and their binding proteins across different macrophages. The data suggest that regulation of glycan-protein complexes may be central to macrophage residence and recruitment.
Collapse
Affiliation(s)
- Diane D Park
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Matthew R Kudelka
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Biochemistry, Emory University, Atlanta, GA 30322, USA
| | - Nan Jia
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Carolyn A Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Revanth Kosaraju
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alykhan M Premji
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Melina Galizzi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Alison V Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Esmaeilzadeh A, Elahi R. Immunobiology and immunotherapy of COVID-19: A clinically updated overview. J Cell Physiol 2021; 236:2519-2543. [PMID: 33022076 PMCID: PMC7675260 DOI: 10.1002/jcp.30076] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new member of the coronavirus family that can cause coronavirus disease 2019 (COVID-19). COVID-9 has become a global pandemic with severe health issues around the world. Identifying the accurate immunopathogenesis of the COVID-19 and the immune response against SARS-CoV-2 is necessary for the development of therapeutic approaches and rational drug design. This paper aims to overview the updated clinical data on the immunopathogenesis of the COVID-19 and review the innate and adaptive immune response to SARS-CoV-2. Also, challenges of the immune response to SARS-CoV-2 leading to dysfunctional immune response and their contribution to the progression of the disease have been discussed. To achieve a more efficient immune response, multiple methods could be applied, including regulation of the immune response, augmentation of the immune system against the virus, inhibition of the dysfunctional immune checkpoints, and inhibition of the viral replication/infection. Based on the immune response against SARS-CoV-2 and its dysfunction, we introduce potential immunotherapies as well as reviewing recruiting/completed clinical trials of COVID-19.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, School of MedicineZanjan University of Medical SciencesZanjanIran
- Immunotherapy Research and Technology GroupZanjan University of Medical SciencesZanjanIran
- Cancer Gene Therapy Research CenterZanjan University of Medical SciencesZanjanIran
| | - Reza Elahi
- Zanjan University of Medical SciencesZanjanIran
| |
Collapse
|
34
|
Miguel CB, da Silva TA, Rodrigues WF, Oliveira-Brito PKM, Roque-Barreira MC, Lazo-Chica JE. Administration of artinm lectin reduces the severity of the acute phase infection with Trypanosoma cruzi. FASEB Bioadv 2021; 3:295-304. [PMID: 33977231 PMCID: PMC8103718 DOI: 10.1096/fba.2020-00065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 11/13/2022] Open
Abstract
The acute phase of experimental Trypanosoma cruzi infection is associated with a strong inflammatory reaction, physiological changes, amastigote nests in tissues, and hematological alterations. ArtinM, a lectin extracted from Artocarpus heterophyllus seeds, is a homotetramer exhibiting immunomodulatory properties that promotes Th1 immune responses against intracellular pathogens, including the induction of neutrophil migration and increase in IL‐12 production. This study aimed to evaluate the effects of ArtinM on experimental Chagas disease in mice. We evaluated mouse survival curves, parasitemia, hematological parameters including quantification of inflammatory infiltrates, and amastigote nests in cardiac tissue during infection. The results showed a reduced number of parasites in the blood, an increase in animal survival, improvements in hematological parameters, and decrease in inflammatory infiltrates and amastigote nests in the group treated with ArtinM. Collectively, these data suggest that the administration of ArtinM can lower the number of parasites in peak parasitemia caused by the Colombian strain of T. cruzi and can increase survival of infected mice. The observed reduction in cardiac tissue injury may be due to fewer T. cruzi amastigote nests and lower levels of inflammatory infiltrates. This study highlights the need for further investigation into the use of ArtinM as a potential alternative therapeutic for treating Chagas disease.
Collapse
Affiliation(s)
- Camila Botelho Miguel
- Postgraduate course in Health Sciences Federal University of Triangulo Mineiro Uberaba Minas Gerais Brazil
| | - Thiago Aparecido da Silva
- Department of Cellular and Molecular Biology and Pathogenic Bioagents Ribeirão Preto Medical School University of São Paulo Ribeirão Preto São Paulo Brazil
| | | | - Patrícia Kellen Martins Oliveira-Brito
- Department of Cellular and Molecular Biology and Pathogenic Bioagents Ribeirão Preto Medical School University of São Paulo Ribeirão Preto São Paulo Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cellular and Molecular Biology and Pathogenic Bioagents Ribeirão Preto Medical School University of São Paulo Ribeirão Preto São Paulo Brazil
| | - Javier Emílio Lazo-Chica
- Postgraduate course in Health Sciences Federal University of Triangulo Mineiro Uberaba Minas Gerais Brazil.,Discipline of Cell Biology/Institute of Biological and Natural Sciences Federal University of Triangulo Mineiro Uberaba Minas Gerais Brazil
| |
Collapse
|
35
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
36
|
Suciati T, Nafisa S, Nareswari TL, Juniatik M, Julianti E, Wibowo MS, Yudhistira T, Ihsanawati I, Triyani Y, Khairurrijal K. ArtinM Grafted Phospholipid Nanoparticles for Enhancing Antibiotic Cellular Uptake Against Intracellular Infection. Int J Nanomedicine 2020; 15:8829-8843. [PMID: 33304099 PMCID: PMC7724644 DOI: 10.2147/ijn.s275449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/25/2020] [Indexed: 11/30/2022] Open
Abstract
Background and Aim An antimicrobial delivery in the form of surface-modified lectin of lipid nanoparticles was proposed to improve cellular accumulation. ArtinM, an active toll-like receptor 2 (TLR2) agonist lectin isolated from cempedak (Arthocarpus integrifolia) seeds, was selected to induce cellular engulfment of nanoparticles within infected host cells. Materials and Methods Lipid nanoparticles were prepared using the emulsification technique before electrostatic adsorption of artinM. The formula comprising of rifampicin, soy phospholipid, and polysorbate 80 was optimized by Box-Behnken design to produce the desired particle size, entrapment efficiency, and drug loading. The optimum formula was characterized for morphology, in vitro release, and cellular transport. Results and Discussion Soy phospholipid showed a profound effect on controlling drug loading and entrapment efficiency. Owing to its surface activity, polysorbate 80 contributed significantly to reduce particle size; however, a higher ratio to lipid concentration resulted in a decrease of rifampicin encapsulation. The adsorption of artinM on the surface of nanoparticles was accomplished by electrostatic binding at pH 4, where this process maintained the stability of encapsulated rifampicin. A high proportion of artinM adsorbed on the surface of the nanoparticles shown by haemagglutination assay, zeta potential measurement, and transmission electron microscopy imaging. Cellular uptake revealed by confocal microscopy showed the success in transporting Nile-red labelled nanoparticles across fibroblast cells. Conclusion The delivery system of nanoparticles bearing artinM becomes a potential platform technology for antibiotic targeting in the treatment of life-threatening chronic diseases caused by intracellular infections.
Collapse
Affiliation(s)
- Tri Suciati
- School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - Safira Nafisa
- Faculty of Pharmacy, Pancasila University, Jakarta, Indonesia
| | | | - Meta Juniatik
- School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - Elin Julianti
- School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | | | - Titah Yudhistira
- Faculty of Industrial Technology, Bandung Institute of Technology, Bandung, Indonesia
| | - Ihsanawati Ihsanawati
- Faculty of Mathematics and Natural Sciences, Bandung Institute of Technology, Bandung, Indonesia
| | - Yani Triyani
- Faculty of Medicine, Bandung Islamic University, Bandung, Indonesia
| | - Khairurrijal Khairurrijal
- Faculty of Mathematics and Natural Sciences, Bandung Institute of Technology, Bandung, Indonesia.,Bioscience and Biotechnology Research Center, Bandung Institute of Technology, Bandung, Indonesia
| |
Collapse
|
37
|
de Campos GY, Oliveira RA, Oliveira-Brito PKM, Roque-Barreira MC, da Silva TA. Pro-inflammatory response ensured by LPS and Pam3CSK4 in RAW 264.7 cells did not improve a fungistatic effect on Cryptococcus gattii infection. PeerJ 2020; 8:e10295. [PMID: 33304649 PMCID: PMC7698691 DOI: 10.7717/peerj.10295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/13/2020] [Indexed: 12/24/2022] Open
Abstract
Background The macrophage lineage is characterized by plasticity due to the acquisition of distinct functional phenotypes, and two major subsets are evaluated; classical M1 activation (strong microbicidal activity) and alternative M2 activation (immunoregulatory functions). The M1 subset expresses inducible nitric oxide synthase (iNOS), which is a primary marker to identify these cells, whereas M2 macrophages are characterized by expression of Arginase-1, found in inflammatory zone 1 (Fizz1), chitinase-like molecule (Ym-1), and CD206. The micro-environmental stimuli and signals in tissues are critical in the macrophage polarization. Toll-like receptors (TLR) ligands, such as lipopolysaccharide (LPS), palmitoyl-3-cysteine-serine-lysine-4 (Pam3CSK4), and ArtinM (mannose-binding lectin) are inductors of M1 subset. The impact of TLR2 and TLR4 signals to fight against Cryptococcus gattii infection is unknown, which is a fungal pathogen that preferentially infects the lung of immunocompetent individuals. The macrophages initiate an immune response to combat the C. gattii, then we evaluated in RAW 264.7 cell the effect of TLR2 and TLR4 agonists on the macrophage polarization dynamic and the impact on the growth of C. gattii. Methods and Results We demonstrated that P3C4, LPS, and ArtinM induced an increase in the levels of iNOS transcripts in RAW 264.7 cells, whereas the relative expression of arginase-1, Ym-1, and Fizz1 was significantly increased in the presence of IL-4 alone. The effects of TLR2 and TLR4 agonists on repolarization from the M2 to M1 subset was evaluated, and the first stimulus was composed of IL-4 and, after 24 h of incubation, the cells were submitted to a second stimulus of P3C4, LPS, ArtinM, or Medium. These TLR agonists induced the production of TNF-α in polarized RAW 264.7 cells to the M2 subset, moreover the measurement of M1/M2 markers using qRT-PCR demonstrated that a second stimulus with LPS for 24 h induced a significant augmentation of levels of iNOS mRNA. This impact of TLR2 and TLR4 agonists in the activation of the RAW 264.7 macrophage was assayed in the presence of C. gattii, the macrophages stimulated with TLR2 and TLR4 agonists for 24 h and co-cultured with C. gattii, as a second stimulus, reached high levels of TNF-α even after incubation with different concentrations of C. gattii. The activation of RAW 264.7 cells induced by TLR2 and TLR4 agonists favored the phagocytosis of C. gattii and inhibited the growth of yeast in the early period of infection. However, RAW 264.7 cells incubated with C. gattii in the presence of TLR2 and TLR4 agonists did not result a significant difference in the colony forming unit (CFU) assay in the early period of C. gattii infection, compared to negative control. Conclusion Polarized RAW 264.7 cells to the M1 subset with TLR2 and TLR4 agonists did not inhibit the growth of C. gattii, whereas robust immunity was identified that could dysregulate host tolerance to this pathogen.
Collapse
Affiliation(s)
- Gabriela Yamazaki de Campos
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Raquel Amorim Oliveira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Patrícia Kellen Martins Oliveira-Brito
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago Aparecido da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
38
|
Nakagawa Y, Yamada S. A novel hypothesis on metal dyshomeostasis and mitochondrial dysfunction in amyotrophic lateral sclerosis: Potential pathogenetic mechanism and therapeutic implications. Eur J Pharmacol 2020; 892:173737. [PMID: 33220280 DOI: 10.1016/j.ejphar.2020.173737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by motor dysfunctions resulting from the loss of upper (UMNs) and lower (LMNs) motor neurons. While ALS symptoms are coincidental with pathological changes in LMNs and UMNs, the causal relationship between the two is unclear. For example, research on the extra-motor symptoms associated with this condition suggests that an imbalance of metals, including copper, zinc, iron, and manganese, is initially induced in the sensory ganglia due to a malfunction of metal binding proteins and transporters. It is proposed that the resultant metal dyshomeostasis may promote mitochondrial dysfunction in the satellite glial cells of these sensory ganglia, causing sensory neuron disturbances and sensory symptoms. Sensory neuron hyperactivation can result in LMN impairments, while metal dyshomeostasis in spinal cord and brain stem parenchyma induces mitochondrial dysfunction in LMNs and UMNs. These events could prompt intracellular calcium dyshomeostasis, pathological TDP-43 formation, and reactive microglia with neuroinflammation, which in turn activate the apoptosis signaling pathways within the LMNs and UMNs. Our model suggests that the degeneration of LMNs and UMNs is incidental to the metal-induced changes in the spinal cord and brain stem. Over time psychiatric symptoms may appear as the metal dyshomeostasis and mitochondrial dysfunction affect other brain regions, including the reticular formation, hippocampus, and prefrontal cortex. It is proposed that metal dyshomeostasis in combination with mitochondrial dysfunction could be the underlying mechanism responsible for the initiation and progression of the pathological changes associated with both the motor and extra-motor symptoms of ALS.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
39
|
Lateral diffusion of CD14 and TLR2 in macrophage plasma membrane assessed by raster image correlation spectroscopy and single particle tracking. Sci Rep 2020; 10:19375. [PMID: 33168941 PMCID: PMC7652837 DOI: 10.1038/s41598-020-76272-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/23/2020] [Indexed: 01/02/2023] Open
Abstract
The diffusion of membrane receptors is central to many biological processes, such as signal transduction, molecule translocation, and ion transport, among others; consequently, several advanced fluorescence microscopy techniques have been developed to measure membrane receptor mobility within live cells. The membrane-anchored receptor cluster of differentiation 14 (CD14) and the transmembrane toll-like receptor 2 (TLR2) are important receptors in the plasma membrane of macrophages that activate the intracellular signaling cascade in response to pathogenic stimuli. The aim of the present work was to compare the diffusion coefficients of CD14 and TLR2 on the apical and basal membranes of macrophages using two fluorescence-based methods: raster image correlation spectroscopy (RICS) and single particle tracking (SPT). In the basal membrane, the diffusion coefficients obtained from SPT and RICS were found to be comparable and revealed significantly faster diffusion of CD14 compared with TLR2. In addition, RICS showed that the diffusion of both receptors was significantly faster in the apical membrane than in the basal membrane, suggesting diffusion hindrance by the adhesion of the cells to the substrate. This finding highlights the importance of selecting the appropriate membrane (i.e., basal or apical) and corresponding method when measuring receptor diffusion in live cells. Accurately knowing the diffusion coefficient of two macrophage receptors involved in the response to pathogen insults will facilitate the study of changes that occur in signaling in these cells as a result of aging and disease.
Collapse
|
40
|
Buranello PAA, Barbosa-Lorenzi VC, Pinto MR, Pereira-da-Silva G, Barreira MCRA, Jamur MC, Oliver C. The lectin ArtinM activates RBL-2H3 mast cells without inducing degranulation. PLoS One 2020; 15:e0230633. [PMID: 32208440 PMCID: PMC7092976 DOI: 10.1371/journal.pone.0230633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/04/2020] [Indexed: 11/19/2022] Open
Abstract
Mast cells are connective tissue resident cells with morphological and functional characteristics that contribute to their role in allergic and inflammatory processes, host defense and maintenance of tissue homeostasis. Mast cell activation results in the release of pro-inflammatory mediators which are largely responsible for the physiological functions of mast cells. The lectin ArtinM, extracted from Artocarpus heterophyllus (jackfruit), binds to D-manose, thus inducing degranulation of mast cells. ArtinM has several immunomodulatory properties including acceleration of wound healing, and induction of cytokine release. The aim of the present study was to investigate the role of ArtinM in the activation and proliferation of mast cells. The rat mast cell line RBL-2H3 was used throughout this study. At a low concentration (0.25μg/mL), ArtinM induced mast cell activation and the release of IL-6 without stimulating the release of pre-formed or newly formed mediators. Additionally, when the cells were activated by ArtinM protein tyrosine phosphorylation was stimulated. The low concentration of ArtinM also activated the transcription factor NFkB, but not NFAT. ArtinM also affected the cell cycle and stimulated cell proliferation. Therefore, ArtinM may have therapeutic applications by modulating immune responses due to its ability to activate mast cells and promote the release of newly synthesized mediators. Additionally, ArtinM could have beneficial effects at low concentrations without degranulating mast cells and inducing allergic reactions.
Collapse
Affiliation(s)
- Patricia A. A. Buranello
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Valéria C. Barbosa-Lorenzi
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcelo R. Pinto
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gabriela Pereira-da-Silva
- Department of Maternal-Infant Nursing and Public Health, Escola de Enfermagem de Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Cristina R. A. Barreira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Célia Jamur
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Constance Oliver
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
41
|
Brouwer MAE, van de Schoor FR, Vrijmoeth HD, Netea MG, Joosten LAB. A joint effort: The interplay between the innate and the adaptive immune system in Lyme arthritis. Immunol Rev 2020; 294:63-79. [PMID: 31930745 PMCID: PMC7065069 DOI: 10.1111/imr.12837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022]
Abstract
Articular joints are a major target of Borrelia burgdorferi, the causative agent of Lyme arthritis. Despite antibiotic treatment, recurrent or persistent Lyme arthritis is observed in a significant number of patients. The host immune response plays a crucial role in this chronic arthritic joint complication of Borrelia infections. During the early stages of B. burgdorferi infection, a major hinder in generating a proper host immune response is the lack of induction of a strong adaptive immune response. This may lead to a delayed hyperinflammatory reaction later in the disease. Several mechanisms have been suggested that might be pivotal for the development of Lyme arthritis and will be highlighted in this review, from molecular mimicry of matrix metallopeptidases and glycosaminoglycans, to autoimmune responses to live bacteria, or remnants of Borrelia spirochetes in joints. Murine studies have suggested that the inflammatory responses are initiated by innate immune cells, but this does not exclude the involvement of the adaptive immune system in this dysregulated immune profile. Genetic predisposition, via human leukocyte antigen-DR isotype and microRNA expression, has been associated with the development of antibiotic-refractory Lyme arthritis. Yet the ultimate cause for (antibiotic-refractory) Lyme arthritis remains unknown. Complex processes of different immune cells and signaling cascades are involved in the development of Lyme arthritis. When these various mechanisms are fully been unraveled, new treatment strategies can be developed to target (antibiotic-refractory) Lyme arthritis more effectively.
Collapse
Affiliation(s)
- Michelle A. E. Brouwer
- Department of Internal MedicineRadboud Center for Infectious Diseases (RCI)Radboud Institute of Molecular Life Sciences (RIMLS)Radboud Institute of Health Sciences (RIHS)Radboud University Medical CenterNijmegenThe Netherlands
| | - Freek R. van de Schoor
- Department of Internal MedicineRadboud Center for Infectious Diseases (RCI)Radboud Institute of Molecular Life Sciences (RIMLS)Radboud Institute of Health Sciences (RIHS)Radboud University Medical CenterNijmegenThe Netherlands
| | - Hedwig D. Vrijmoeth
- Department of Internal MedicineRadboud Center for Infectious Diseases (RCI)Radboud Institute of Molecular Life Sciences (RIMLS)Radboud Institute of Health Sciences (RIHS)Radboud University Medical CenterNijmegenThe Netherlands
| | - Mihai G. Netea
- Department of Internal MedicineRadboud Center for Infectious Diseases (RCI)Radboud Institute of Molecular Life Sciences (RIMLS)Radboud Institute of Health Sciences (RIHS)Radboud University Medical CenterNijmegenThe Netherlands
- Department for Genomics & ImmunoregulationLife and Medical Sciences Institute (LIMES)University of BonnBonnGermany
| | - Leo A. B. Joosten
- Department of Internal MedicineRadboud Center for Infectious Diseases (RCI)Radboud Institute of Molecular Life Sciences (RIMLS)Radboud Institute of Health Sciences (RIHS)Radboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
42
|
Abstract
Complex diseases involve dynamic perturbations of pathophysiological processes during disease progression. Transcriptional programs underlying such perturbations are unknown in many diseases. Here, we present core transcriptional regulatory circuits underlying early and late perturbations in prion disease. We first identified cellular processes perturbed early and late using time-course gene expression data from three prion-infected mouse strains. We then built a transcriptional regulatory network (TRN) describing regulation of early and late processes. We found over-represented feed-forward loops (FFLs) comprising transcription factor (TF) pairs and target genes in the TRN. Using gene expression data of brain cell types, we further selected active FFLs where TF pairs and target genes were expressed in the same cell type and showed correlated temporal expression changes in the brain. We finally determined core transcriptional regulatory circuits by combining these active FFLs. These circuits provide insights into transcriptional programs for early and late pathophysiological processes in prion disease.
Collapse
|
43
|
SNP Diversity in CD14 Gene Promoter Suggests Adaptation Footprints in Trypanosome Tolerant N'Dama ( Bos taurus) but not in Susceptible White Fulani ( Bos indicus) Cattle. Genes (Basel) 2020; 11:genes11010112. [PMID: 31963925 PMCID: PMC7017169 DOI: 10.3390/genes11010112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
Immune response to infections has been shown to be mediated by genetic diversity in pattern recognition receptors, leading to disease tolerance or susceptibility. We elucidated naturally occurring variations within the bovine CD14 gene promoter in trypanosome-tolerant (N'Dama) and susceptible (White Fulani) cattle, with genomic and computational approaches. Blood samples were collected from White Fulani and N'Dama cattle, genomic DNA extracted and the entire promoter region of the CD14 gene amplified by PCR. We sequenced this region and performed in silico computation to identify SNP variants, transcription factor binding sites, as well as micro RNAs in the region. CD14 promoter sequences were compared with the reference bovine genome from the Ensembl database to identify various SNPs. Furthermore, we validated three selected N'Dama specific SNPs using custom Taqman SNP genotyping assay for genetic diversity. In all, we identified a total of 54 and 41 SNPs at the CD14 promoter for N'Dama and White Fulani respectively, including 13 unique SNPs present in N'Dama only. The significantly higher SNP density at the CD14 gene promoter region in N'Dama may be responsible for disease tolerance, possibly an evolutionary adaptation. Our genotype analysis of the three loci selected for validation show that mutant alleles (A/A, C/C, and A/A) were adaptation profiles within disease tolerant N'Dama. A similar observation was made for our haplotype analysis revealing that haplotypes H1 (ACA) and H2 (ACG) were significant combinations within the population. The SNP effect prediction revealed 101 and 89 new transcription factor binding sites in N'Dama and White Fulani, respectively. We conclude that disease tolerant N'Dama possessing higher SNP density at the CD14 gene promoter and the preponderance of mutant alleles potentially confirms the significance of this promoter in immune response, which is lacking in susceptible White Fulani. We, therefore, recommend further in vitro and in vivo study of this observation in infected animals, as the next step for understanding genetic diversity relating to varying disease phenotypes in both breeds.
Collapse
|
44
|
Transcriptomic Profiles of Monocyte-Derived Macrophages in Response to Escherichia coli is Associated with the Host Genetics. Sci Rep 2020; 10:271. [PMID: 31937813 PMCID: PMC6959288 DOI: 10.1038/s41598-019-57089-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/21/2019] [Indexed: 01/05/2023] Open
Abstract
Reactive Nitrogen Species (RNS) are a group of bactericidal molecules produced by macrophages in response to pathogens in a process called oxidative burst. Nitric oxide (NO-) is a member of RNS produced from arginine by inducible Nitric Oxide Synthase (iNOS) enzyme. The activity of iNOS and production of NO- by macrophages following stimulation is one of the indicators of macrophage polarization towards M1/proinflammatory. Production of NO- by bovine monocyte-derived macrophage (MDM) and mouse peritoneal macrophages has been shown to be strongly associated with host genetic with the heritability of 0.776 in bovine MDM and 0.8 in mouse peritoneal macrophages. However, the mechanism of genetic regulation of macrophage response has remained less explored. In the current study, the transcriptome of bovine MDMs was compared between two extreme phenotypes that had been classified as high and low responder based on NO- production. The results showed that 179 and 392 genes were differentially expressed (DE) between high and low responder groups at 3 and 18 hours after exposure to Escherichia coli, respectively. A set of 11 Transcription Factors (TFs) (STAT1, IRF7, SPI1, STAT4, IRF1, HIF1A, FOXO3, REL, NFAT5, HIC1, and IRF4) at 3 hours and a set of 13 TFs (STAT1, IRF1, HIF1A, STAT4, ATF4, TP63, EGR1, CDKN2A, RBL1, E2F1, PRDM1, GATA3, and IRF4) at 18 hours after exposure to E. coli were identified to be differentially regulated between the high and low responder phenotypes. These TFs were found to be divided into two clusters of inflammatory- and hypoxia-related TFs. Functional analysis revealed that some key canonical pathways such as phagocytosis, chemotaxis, antigen presentation, and cell-to-cell signalling are enriched among the over-expressed genes by high responder phenotype. Based on the results of this study, it was inferred that the functional characteristics of bovine MDMs are associated with NO-based classification. Since NO- production is strongly associated with host genetics, this study for the first time shows the distinct proinflammatory profiles of macrophages are controlled by the natural genetic polymorphism in an outbred population. In addition, the results suggest that genetics can be considered as a new dimension in the current model of macrophage polarization which is currently described by the combination of stimulants, only.
Collapse
|
45
|
da Silva TA, Oliveira-Brito PKM, de Oliveira Thomaz SM, Roque-Barreira MC. ArtinM: Purification and Evaluation of Biological Activities. Methods Mol Biol 2020; 2132:349-358. [PMID: 32306342 DOI: 10.1007/978-1-0716-0430-4_34] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The immunomodulatory activity of plant lectins has been evaluated because of their high selectivity for glycans linked to receptors on innate and adaptative immune cells. ArtinM is a mannosyl-binding lectin, obtained from the seeds of Artocarpus heterophyllus, that induces the differentiation of CD4+ T cells and macrophages by interacting with CD3 and TLR2/CD14, respectively. This ArtinM property ultimately favors the combat of intracellular pathogens, opening new perspectives on the lectins application as immunomodulatory agents. The current section describes protocols for purification and evaluation of ArtinM biological activity. The purification is based on the ArtinM-D-mannose affinity. The effect of inducing IL-12 production by murine macrophages cell line is adopted to evaluate the ArtinM biological activity.
Collapse
Affiliation(s)
- Thiago Aparecido da Silva
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Patrícia Kellen Martins Oliveira-Brito
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Sandra Maria de Oliveira Thomaz
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria Cristina Roque-Barreira
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine University of São Paulo (FMRP/USP), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
46
|
Costa Mendonça-Natividade F, Duque Lopes C, Ricci-Azevedo R, Sardinha-Silva A, Figueiredo Pinzan C, Paiva Alegre-Maller AC, L Nohara L, B Carneiro A, Panunto-Castelo A, C Almeida I, Roque-Barreira MC. Receptor Heterodimerization and Co-Receptor Engagement in TLR2 Activation Induced by MIC1 and MIC4 from Toxoplasma gondii. Int J Mol Sci 2019; 20:ijms20205001. [PMID: 31658592 PMCID: PMC6829480 DOI: 10.3390/ijms20205001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 01/07/2023] Open
Abstract
The microneme organelles of Toxoplasma gondii tachyzoites release protein complexes (MICs), including one composed of the transmembrane protein MIC6 plus MIC1 and MIC4. In this complex, carbohydrate recognition domains of MIC1 and MIC4 are exposed and interact with terminal sialic acid and galactose residues, respectively, of host cell glycans. Recently, we demonstrated that MIC1 and MIC4 binding to the N-glycans of Toll-like receptor (TLR) 2 and TLR4 on phagocytes triggers cell activation and pro-inflammatory cytokine production. Herein, we investigated the requirement for TLR2 heterodimerization and co-receptors in MIC-induced responses, as well as the signaling molecules involved. We used MICs to stimulate macrophages and HEK293T cells transfected with TLR2 and TLR1 or TLR6, both with or without the co-receptors CD14 and CD36. Then, the cell responses were analyzed, including nuclear factor-kappa B (NF-κB) activation and cytokine production, which showed that (1) only TLR2, among the studied factors, is crucial for MIC-induced cell activation; (2) TLR2 heterodimerization augments, but is not critical for, activation; (3) CD14 and CD36 enhance the response to MIC stimulus; and (4) MICs activate cells through a transforming growth factor beta-activated kinase 1 (TAK1)-, mammalian p38 mitogen-activated protein kinase (p38)-, and NF-κB-dependent pathway. Remarkably, among the studied factors, the interaction of MIC1 and MIC4 with TLR2 N-glycans is sufficient to induce cell activation, which promotes host protection against T. gondii infection.
Collapse
Affiliation(s)
- Flávia Costa Mendonça-Natividade
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| | - Carla Duque Lopes
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| | - Rafael Ricci-Azevedo
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| | - Aline Sardinha-Silva
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| | - Camila Figueiredo Pinzan
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| | - Ana Claudia Paiva Alegre-Maller
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| | - Lilian L Nohara
- Border Biomedical Research Center (BBRC), Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX 79968, USA.
| | - Alan B Carneiro
- Border Biomedical Research Center (BBRC), Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX 79968, USA.
- Institute of Medical Biochemistry, Program of Molecular Biology and Biotechnology at Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro RJ 21941-599, Brazil.
| | - Ademilson Panunto-Castelo
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo USP (FFCLRP/USP), Ribeirão Preto SP 14040-900, Brazil.
| | - Igor C Almeida
- Border Biomedical Research Center (BBRC), Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX 79968, USA.
| | - Maria Cristina Roque-Barreira
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirão Preto SP 14049-900, Brazil.
| |
Collapse
|
47
|
Malvandi AM, Loretelli C, Ben Nasr M, Zuccotti GV, Fiorina P. Sitagliptin favorably modulates immune-relevant pathways in human beta cells. Pharmacol Res 2019; 148:104405. [DOI: 10.1016/j.phrs.2019.104405] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/13/2019] [Accepted: 08/19/2019] [Indexed: 10/26/2022]
|
48
|
Oliveira-Brito PKM, Rezende CP, Almeida F, Roque-Barreira MC, da Silva TA. iNOS/Arginase-1 expression in the pulmonary tissue over time during Cryptococcus gattii infection. Innate Immun 2019; 26:117-129. [PMID: 31446837 PMCID: PMC7016403 DOI: 10.1177/1753425919869436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inhalation of Cryptococcus gattii yeasts (causing cryptococcosis) triggers an anti-cryptococcal immune response initiated by macrophages, neutrophils or dendritic cells, and the iNOS expressed by various cells may regulate the function and differentiation of innate and adaptive immune cells. Here, we evaluated the effect of progression of C. gattii infection on the host innate immune response. C. gattii infection in BALB/c mice spreads to several organs by 21 d post infection. The numbers of neutrophils and lymphocytes in the peripheral blood of C. gattii–infected mice were remarkably altered on that day. The frequency of CD11b+ cells and cell concentrations of CD4+ and CD8+ T cells was significantly altered in the pulmonary tissue of infected mice. We found a higher frequency of CD11b+/iNOS+ cells in the lungs of infected mice, accompanied by an increase in frequency of CD11b+/Arginase-1+ cells over time. Moreover, the iNOS/Arginase-1 expression ratio in CD11b+ cells reached its lowest value at 21 d post infection. In addition, the cytokine micro-environment in infected lungs did not show a pro-inflammatory profile. Surprisingly, iNOS knock-out prolonged the survival of infected mice, while their pulmonary fungal burden was higher than that of infected WT mice. Thus, C. gattii infection alters the immune response in the pulmonary tissue, and iNOS expression may play a key role in infection progression.
Collapse
Affiliation(s)
- Patrícia Kellen Martins Oliveira-Brito
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Brazil.,These authors contributed equally to this work
| | - Caroline Patini Rezende
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Brazil.,These authors contributed equally to this work
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Thiago Aparecido da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
49
|
de Sousa JR, Da Costa Vasconcelos PF, Quaresma JAS. Functional aspects, phenotypic heterogeneity, and tissue immune response of macrophages in infectious diseases. Infect Drug Resist 2019; 12:2589-2611. [PMID: 31686866 PMCID: PMC6709804 DOI: 10.2147/idr.s208576] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are a functionally heterogeneous group of cells with specialized functions depending not only on their subgroup but also on the function of the organ or tissue in which the cells are located. The concept of macrophage phenotypic heterogeneity has been investigated since the 1980s, and more recent studies have identified a diverse spectrum of phenotypic subpopulations. Several types of macrophages play a central role in the response to infectious agents and, along with other components of the immune system, determine the clinical outcome of major infectious diseases. Here, we review the functions of various macrophage phenotypic subpopulations, the concept of macrophage polarization, and the influence of these cells on the evolution of infections. In addition, we emphasize their role in the immune response in vivo and in situ, as well as the molecular effectors and signaling mechanisms used by these cells. Furthermore, we highlight the mechanisms of immune evasion triggered by infectious agents to counter the actions of macrophages and their consequences. Our aim here is to provide an overview of the role of macrophages in the pathogenesis of critical transmissible diseases and discuss how elucidation of this relationship could enhance our understanding of the host-pathogen association in organ-specific immune responses.
Collapse
Affiliation(s)
- Jorge Rodrigues de Sousa
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
| | - Pedro Fernando Da Costa Vasconcelos
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
| | - Juarez Antonio Simões Quaresma
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
- School of Medicine, São Paulo University, São Paulo, SP, Brazil
| |
Collapse
|
50
|
Trindade R, Albrektsson T, Galli S, Prgomet Z, Tengvall P, Wennerberg A. Bone Immune Response to Materials, Part II: Copper and Polyetheretherketone (PEEK) Compared to Titanium at 10 and 28 Days in Rabbit Tibia. J Clin Med 2019; 8:jcm8060814. [PMID: 31181635 PMCID: PMC6616385 DOI: 10.3390/jcm8060814] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022] Open
Abstract
Osseointegration is likely the result of an immunologically driven bone reaction to materials such as titanium. Osseointegration has resulted in the clinical possibility to anchor oral implants in jaw bone tissue. However, the mechanisms behind bony anchorage are not fully understood and complications over a longer period of time have been reported. The current study aims at exploring possible differences between copper (Cu) and polyetheretherketone (PEEK) materials that do not osseointegrate, with osseointegrating cp titanium as control. The implants were placed in rabbit tibia and selected immune markers were evaluated at 10 and 28 days of follow-up. Cu and PEEK demonstrated at both time points a higher immune activation than cp titanium. Cu demonstrated distance osteogenesis due to a maintained proinflammatory environment over time, and PEEK failed to osseointegrate due to an immunologically defined preferential adipose tissue formation on its surface. The here presented results suggest the description of two different mechanisms for failed osseointegration, both of which are correlated to the immune system.
Collapse
Affiliation(s)
- Ricardo Trindade
- Department of Prosthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Tomas Albrektsson
- Department of Biomaterials, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden.
- Department of Prosthodontics, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Silvia Galli
- Department of Prosthodontics, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Zdenka Prgomet
- Department of Oral Pathology, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Pentti Tengvall
- Department of Biomaterials, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Ann Wennerberg
- Department of Prosthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|