1
|
Tang M, Li H, Chang S, Li Y, Nie H, Li F. Dysregulated circular RNAs in rheumatoid arthritis: Cellular roles and clinical prospects. Autoimmun Rev 2025:103774. [PMID: 39956349 DOI: 10.1016/j.autrev.2025.103774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Rheumatoid arthritis (RA) is still a healthcare challenge, although current therapeutic strategies have substantially improved its clinical outcomes. The development of novel biomarkers and treatments can increase the likelihood of identification and disease remission in RA patients, especially for patients with seronegative RA and difficult-to-treat RA (D2T RA). Circular RNAs (circRNAs), a novel non-coding RNA species, have been reported to play crucial roles in various biological process of RA. The mechanistic functions of the dysregulated circRNAs in RA are primarily associated with miRNA sponging and regulating transcription. CircRNAs acting as miRNA sponges are further summarized by cell types, including fibroblast-like synoviocytes (FLSs), lymphocytes, macrophages, chondrocytes, and mesenchymal stem cells (MSCs)-/plasma-secreted exosomes. Besides, a description of dysregulated circRNAs in blood, synovial tissue and cartilage tissue suggests their diagnostic potential for RA. In addition, some directions for future research are provided to open the possibility that dysregulated cell- and tissue- specific circRNAs constituting a fresh reservoir of therapeutic targets, and biomarkers for diagnosis, predicting response to therapy, drug selection or patient stratification for RA.
Collapse
Affiliation(s)
- Mengshi Tang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research for Systemic Autoimmune Disease in Hunan Province, 410011, China
| | - Hongxing Li
- Department of Orthopaedics, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Orthopaedics, The Central Hospital of Shaoyang, Shaoyang, Hunan 422099, China
| | - Siyuan Chang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research for Systemic Autoimmune Disease in Hunan Province, 410011, China
| | - Yuanyuan Li
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research for Systemic Autoimmune Disease in Hunan Province, 410011, China
| | - Huiyu Nie
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research for Systemic Autoimmune Disease in Hunan Province, 410011, China
| | - Fen Li
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Clinical Medical Research for Systemic Autoimmune Disease in Hunan Province, 410011, China.
| |
Collapse
|
2
|
Chayé MAM, van Hengel ORJ, Voskamp AL, Ozir-Fazalalikhan A, König MH, Stam KA, Manurung MD, Mouwenda YD, Aryeetey YA, Kurniawan A, Kruize YCM, Sartono E, Buisman AM, Yazdanbakhsh M, Tak T, Smits HH. Multi-dimensional analysis of B cells reveals the expansion of memory and regulatory B-cell clusters in humans living in rural tropical areas. Clin Exp Immunol 2025; 219:uxae074. [PMID: 39129562 PMCID: PMC11771192 DOI: 10.1093/cei/uxae074] [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: 10/09/2023] [Revised: 07/06/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024] Open
Abstract
B-cells play a critical role in the formation of immune responses against pathogens by acting as antigen-presenting cells, by modulating immune responses, and by generating immune memory and antibody responses. Here, we studied B-cell subset distributions between regions with higher and lower microbial exposure, i.e. by comparing peripheral blood B-cells from people living in Indonesia or Ghana to those from healthy Dutch residents using a 36-marker mass cytometry panel. By applying an unbiased multidimensional approach, we observed differences in the balance between the naïve and memory compartments, with higher CD11c+ and double negative (DN-IgDnegCD27neg) memory (M)B-cells in individuals from rural tropical areas, and conversely lower naïve B-cells compared to residents from an area with less pathogen exposure. Furthermore, characterization of total B-cell populations, CD11c+, DN, and Breg cells showed the emergence of specific memory clusters in individuals living in rural tropical areas. Some of these differences were more pronounced in children compared to adults and suggest that a higher microbial exposure accelerates memory B-cell formation, which "normalizes" with age.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Oscar R J van Hengel
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Astrid L Voskamp
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | | | - Marion H König
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Koen A Stam
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Mikhael D Manurung
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Yoanne D Mouwenda
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Yvonne A Aryeetey
- Parasitology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Agnes Kurniawan
- Department of Parasitology, Universitas Indonesia, Jakarta, Indonesia
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Erliyani Sartono
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Anne-Marie Buisman
- Laboratory for Immunology of Infectious Diseases and Vaccines, Center for Infectious Diseases Control, National Institute for Public Health and The Environment, Bilthoven, The Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Tamar Tak
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Hermelijn H Smits
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| |
Collapse
|
3
|
Zhang Y, Qin X, Lou W, Wang L, Lu W, Gao C, Hu S. Deciphering the cellular landscape and potential targets of nasopharyngeal and oral cancers using single-cell RNA sequencing. Cell Biol Int 2024; 48:1849-1861. [PMID: 39205595 DOI: 10.1002/cbin.12236] [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: 02/08/2024] [Revised: 07/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Cellular heterogeneity in nasopharyngeal cancer (NPC) and oral cancer remains unclear. In the current study, using single-cell RNA sequencing techniques, we investigated the cellular landscape in NPC and oral cancers. We identified a diverse range of cell types within the tumor microenvironment (TME) and variations in cell infiltration between NPC and oral cancer. In oral cancer, we observed a predominant infiltration of epithelial cells, fibroblasts, and endothelial cells (ECs), while T cells were the main infiltrating cell population in NPCs. We further classified these infiltrating cells into subclusters. Additionally, we observed complex interactions among cells that led to distinct trajectories. In particular, a unique epithelial subcluster with high expression of major histocompatibility complex class II (MHC-II) molecules was correlated with a favorable outcome and infiltration of CD4+ T cells. In addition, MHC-II+ epithelial cells inhibited mouse tumor growth and promoted T-cell infiltration. Consequently, our findings provide a deep understanding of the TME showing a significant prognostic value and therapeutic potential.
Collapse
Affiliation(s)
- Yanfei Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyu Qin
- Department of Vascular Surgery, Zhengzhou Central Hospital, Zhengzhou, Henan, China
| | - Weihua Lou
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wuhao Lu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Changhui Gao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shousen Hu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Liguori A, D'Ambrosio F, Napodano C, Gentili V, Giustiniani MC, Pompili M, Grieco A, Rapaccini G, Urbani A, Gasbarrini A, Basile U, Miele L. Serum-free light chains as a dependable biomarker for stratifying patients with metabolic dysfunction-associated steatotic liver disease. Liver Int 2024; 44:2625-2638. [PMID: 39016540 DOI: 10.1111/liv.16036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/29/2024] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND AND AIMS Adaptive immunity is gaining a significant role in progression of metabolic dysfunction-associated steatotic liver disease (MASLD). B-cell activity can be assessed by serum-free light chains (sFLCs) k and λ levels. The objective of the present investigation is to examine the utility of sFLCs as non-invasive biomarkers for the stratification of MASLD. METHODS We enrolled a consecutive cohort from an outpatient liver unit. Diagnosis of metabolic dysfunction-associated steatohepatitis (MASH) was made with liver biopsy according to current guidelines. Compensated advanced chronic liver disease (cACLD) and clinically significant portal hypertension (CSPH) were defined according to Baveno VII criteria. sFLCs were measured by turbidimetry using an immunoassay. RESULTS We evaluated 254 patients, 162/254 (63.8%) were male. Median age was 54 years old, and the median body mass index was 28.4 kg/m2. A total of 157/254 (61.8%) subjects underwent liver biopsy: 88 had histological diagnosis of MASH, 89 were considered as simple metabolic dysfunction-associated steatotic liver (MASL) and 77/254 (30.3%) patients with compensated metabolic dysfunction-associated cirrhosis. By using Baveno VII criteria, 101/254 (39.7%) patients had cACLD; among them, 45/101 (44.5%) had CSPH. Patients with cACLD showed higher sFLC levels compared with patients without cACLD (p < .01), and patients with CSPH showed higher sFLC levels than patients without CSPH (p < .01). At multivariable analysis, sFLCs were associated with cACLD (p < .05) independently from γ-globulins and other known dysmetabolic risk factors. κFLC was associated with CSPH (p < .05) independently from γ-globulins and other known dysmetabolic risk factors. CONCLUSION sFLCs could be a simple biomarker for stratification of cACLD in MASLD patients.
Collapse
Affiliation(s)
- Antonio Liguori
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Internal Medicine and Liver Transplant Unit, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
- CEMAD, Digestive Disease Center, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Francesca D'Ambrosio
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cecilia Napodano
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanessa Gentili
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Cristina Giustiniani
- Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Maurizio Pompili
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Grieco
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gianludovico Rapaccini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Urbani
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Internal Medicine and Liver Transplant Unit, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti HospitalAUSL Latina, Latina, Italy
| | - Luca Miele
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Internal Medicine and Liver Transplant Unit, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
- CEMAD, Digestive Disease Center, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| |
Collapse
|
5
|
Ransegnola BP, Pattarabanjird T, McNamara CA. Tipping the Scale: Atheroprotective IgM-Producing B Cells in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1906-1915. [PMID: 39022832 PMCID: PMC11338718 DOI: 10.1161/atvbaha.124.319847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease whose progression is fueled by proinflammatory moieties and limited by anti-inflammatory mediators. Whereas oxidative damage and the generation of oxidation-specific epitopes that act as damage-associated molecular patterns are highly inflammatory, IgM antibodies produced by B-1 and marginal zone B cells counteract unrestricted inflammation by neutralizing and encouraging clearance of these proinflammatory signals. In this review, we focus on describing the identities of IgM-producing B cells in both mice and humans, elaborating the mechanisms underlying IgM production, and discussing the potential strategies to augment the production of atheroprotective IgM. In addition, we will discuss promising therapeutic interventions in humans to help tip the scale toward augmentation of IgM production and to provide atheroprotection.
Collapse
Affiliation(s)
- Brett Patrick Ransegnola
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Pathology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tanyaporn Pattarabanjird
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Coleen A. McNamara
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
6
|
Fleige L, Fillatreau S, Claus M, Capellino S. Additional use of α-IgM antibodies potentiates CpG ODN2006-induced B cell activation by targeting mainly naïve and marginal zone-like B cells. Cell Immunol 2024; 403-404:104846. [PMID: 38996539 DOI: 10.1016/j.cellimm.2024.104846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024]
Abstract
CpG ODN2006 is widely used as a potent B cell stimulant in vitro and in vivo. However, it shows a deficit in targeting naïve B cells in vitro. In this study, we investigated whether α-IgM can support ODN2006-induced effects on B cells to obtain enhanced activation with focus on different B cell subsets. Our results delineated robust B cell activation, shown by increased activation marker expression and cytokine secretion by each agent alone, and further augmented when used in combination. Interestingly, α-IgM targeted mainly naïve and marginal zone-like B cells, thus complementing the pronounced effects of ODN2006 on memory B cells and achieving optimal activation for all B cell subsets. Taken together, combining ODN2006 and α-IgM is beneficial for in vitro activation including all B cell subsets. Furthermore, our results suggest that α-IgM could enhance efficacy of ODN2006 in vivo with further need of investigation.
Collapse
Affiliation(s)
- Leonie Fleige
- Department of Immunology, Research Group of Neuroimmunology, IfADo-Leibniz Research Centre for Working Environment and Human Factors, Ardeystraße 67, 44139 Dortmund, Germany.
| | - Simon Fillatreau
- Department of Immunology, Infectiology and Haematology, Research Group of Immunity in health and disease, Institut Necker Enfants Malades, Faculté de Médecine Necker, 160 rue de Vaugirard, 75015 Paris Cedex, France
| | - Maren Claus
- Department of Immunology, Research Group of Immunmodulation, IfADo-Leibniz Research Centre for Working Environment and Human Factors, Ardeystraße 67, 44139 Dortmund, Germany
| | - Silvia Capellino
- Department of Immunology, Research Group of Neuroimmunology, IfADo-Leibniz Research Centre for Working Environment and Human Factors, Ardeystraße 67, 44139 Dortmund, Germany
| |
Collapse
|
7
|
Fukasawa T, Yoshizaki-Ogawa A, Sato S, Yoshizaki A. The role of B cells in systemic sclerosis. J Dermatol 2024; 51:904-913. [PMID: 38321641 DOI: 10.1111/1346-8138.17134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/08/2024]
Abstract
Systemic sclerosis (SSc) is a rare and refractory systemic disease characterized by fibrosis and vasculopathy in the presence of autoimmune abnormalities. While the exact cause of SSc is incompletely understood, the specific autoantibodies identified in SSc are closely linked to disease severity and prognosis, indicating a significant role of autoimmune abnormalities in the pathogenesis of SSc. Although the direct pathogenic mechanisms of autoantibodies in SSc are not fully elucidated, numerous prior investigations have demonstrated the involvement of B cells in the pathogenesis of SSc through various mechanisms. Additionally, several clinical trials have explored the efficacy of B-cell depletion therapy for SSc, with many reporting positive outcomes. However, the role of B cells in SSc pathogenesis is multifaceted, as they can both promote inflammation and exert inhibitory functions. This article provides an overview of the involvement of B cells in SSc development, incorporating the latest research findings.
Collapse
Affiliation(s)
- Takemichi Fukasawa
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Rasool D, Jan SA, Khan SU, Nahid N, Ashfaq UA, Umar A, Qasim M, Noor F, Rehman A, Shahzadi K, Alshammari A, Alharbi M, Nisar MA. Subtractive proteomics-based vaccine targets annotation and reverse vaccinology approaches to identify multiepitope vaccine against Plesiomonas shigelloides. Heliyon 2024; 10:e31304. [PMID: 38845922 PMCID: PMC11153098 DOI: 10.1016/j.heliyon.2024.e31304] [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: 03/30/2023] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024] Open
Abstract
Plesiomonas shigelloides, an aquatic bacterium belonging to the Enterobacteriaceae family, is a frequent cause of gastroenteritis with diarrhea and gastrointestinal severe disease. Despite decades of research, discovering a licensed and globally accessible vaccine is still years away. Developing a putative vaccine that can combat the Plesiomonas shigelloides infection by boosting population immunity against P. shigelloides is direly needed. In the framework of the current study, the entire proteome of P. shigelloides was explored using subtractive genomics integrated with the immunoinformatics approach for designing an effective vaccine construct against P. shigelloides. The overall stability of the vaccine construct was evaluated using molecular docking, which demonstrated that MEV showed higher binding affinities with toll-like receptors (TLR4: 51.5 ± 10.3, TLR2: 60.5 ± 9.2) and MHC receptors(MHCI: 79.7 ± 11.2 kcal/mol, MHCII: 70.4 ± 23.7). Further, the therapeutic efficacy of the vaccine construct for generating an efficient immune response was evaluated by computational immunological simulation. Finally, computer-based cloning and improvement in codon composition without altering amino acid sequence led to the development of a proposed vaccine. In a nutshell, the findings of this study add to the existing knowledge about the pathogenesis of this infection. The schemed MEV can be a possible prophylactic agent for individuals infected with P. shigelloides. Nevertheless, further authentication is required to guarantee its safeness and immunogenic potential.
Collapse
Affiliation(s)
- Danish Rasool
- Department of Bioinformatics and Biosciences, Capital University of Science and Technology, Islamabad, 44000, Pakistan
| | - Sohail Ahmad Jan
- Department of Bioinformatics and Biosciences, Capital University of Science and Technology, Islamabad, 44000, Pakistan
| | | | - Nazia Nahid
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Ahitsham Umar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Fatima Noor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A & F Uiversity, yangling, 712100, Shaanxi, China
| | - Kiran Shahzadi
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Muhammad Atif Nisar
- College of Science and Engineering, Flinders University, Adelaide, 5042, Australia
| |
Collapse
|
9
|
Yuan C, Lin Y, Wang Y, Zhang Y, Zhao X, Yuan H, Li T, Song Q. Effects of porcine epidemic diarrhea virus infection on CD21 + B cells activation. Vet Microbiol 2024; 293:110087. [PMID: 38663176 DOI: 10.1016/j.vetmic.2024.110087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 05/15/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a devastating pathogen of acute- gastrointestinal infectious diseases, which can cause vomiting, diarrhea, dehydration and high morbidity and mortality among neonatal piglets. Humoral immunity plays a vital role in the host anti-PEDV infection process, but the mechanism of PEDV-induced B-cell immune response remains unknown. In this study, the effects of PEDV infection on CD21+ B cell activation were systematically analyzed through animal experiments. Enzyme-linked immunosorbent assays (ELISA) revealed that low levels of serum-specific IgA, IgM, or IgG were detected in piglets after PEDV infection, respectively. Serum interleukin (IL)-6 levels increased significantly at 4 d after infection, and the levels of IL-4, B-cell activating factor (BAFF), interferon (IFN)-γ, transforming growth factor (TGF)-β and IL-10 decreased at 7 d after infection. Fluorescence-activated cell sorting (FACS) showed that expression levels of CD21, MHC Ⅱ, CD40, and CD38 on B cell surfaces were significantly higher. In contrast, the proportions of CD21+IgM+ B cells were decreased in peripheral blood mononuclear cells (PBMCs) from the infected piglets. No differences were found in the percentage of CD21+CD80+ and CD21+CD27+ B cells in PBMCs from the infected piglets. In addition, the number of CD21+B cells in PBMCs stimulated with PEDV in vitro was significantly lower. No significant change in the mRNA expression of BCR molecules was found while the expression levels of paired immunoglobulin-like receptor B (PIR-B), B cell adaptor molecule of 32 kDa (Bam32) and BAFF were decreased. In conclusion, our research demonstrates that virulent strains of PEDV profoundly impact B cell activation, leading to alterations in phenotypic expression and BCR signaling molecules. Furthermore, this dysregulation results in compromised specific antibody secretion and perturbed cytokine production, highlighting the intricate immunological dysfunctions induced by PEDV infection.
Collapse
Affiliation(s)
- Chen Yuan
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yidan Lin
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yawen Wang
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yanan Zhang
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Xue Zhao
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Hongxing Yuan
- Agriculture and Rural Bureau of Guantao County, Handan, Hebei Province 057750, China
| | - Tanqing Li
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Qinye Song
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China.
| |
Collapse
|
10
|
Elias C, Chen C, Cherukuri A. Regulatory B Cells in Solid Organ Transplantation: From Immune Monitoring to Immunotherapy. Transplantation 2024; 108:1080-1089. [PMID: 37779239 PMCID: PMC10985051 DOI: 10.1097/tp.0000000000004798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Regulatory B cells (Breg) modulate the immune response in diverse disease settings including transplantation. Despite the lack of a specific phenotypic marker or transcription factor, their significance in transplantation is underscored by their ability to prolong experimental allograft survival, the possibility for their clinical use as immune monitoring tools, and the exciting prospect for them to form the basis for cell therapy. Interleukin (IL)-10 expression remains the most widely used marker for Breg. Several Breg subsets with distinct phenotypes that express this "signature Breg cytokine" have been described in mice and humans. Although T-cell immunoglobulin and mucin family-1 is the most inclusive and functional marker that accounts for murine Breg with disparate mechanisms of action, the significance of T-cell immunoglobulin and mucin family-1 as a marker for Breg in humans still needs to be explored. Although the primary focus of this review is the role of Breg in clinical transplantation, the net modulatory effect of B cells on the immune response and clinical outcomes is the result of the balancing functions of both Breg and effector B cells. Supporting this notion, B-cell IL-10/tumor necrosis factor α ratio is shown to predict immunologic reactivity and clinical outcomes in kidney and liver transplantation. Assessment of Breg:B effector balance using their IL-10/tumor necrosis factor α ratio may identify patients that require more immunosuppression and provide mechanistic insights into potential therapies. In summary, current advances in our understanding of murine and human Breg will pave way for future definitive clinical studies aiming to test them for immune monitoring and as therapeutic targets.
Collapse
Affiliation(s)
- Charbel Elias
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chuxiao Chen
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Aravind Cherukuri
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Renal and Electrolyte Division, Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Petinati NA, Sadovskaya AV, Sats NV, Kapranov NM, Davydova YO, Fastova EA, Magomedova AU, Vasilyeva AN, Aleshina OA, Arapidi GP, Shender VO, Smirnov IP, Pobeguts OV, Lagarkova MA, Drize NI, Parovichnikova EN. Molecular Changes in Immunological Characteristics of Bone Marrow Multipotent Mesenchymal Stromal Cells in Lymphoid Neoplasia. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:883-903. [PMID: 38880649 DOI: 10.1134/s0006297924050092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 06/18/2024]
Abstract
Immune system and bone marrow stromal cells play an important role in maintaining normal hematopoiesis. Lymphoid neoplasia disturbs not only development of immune cells, but other immune response mechanisms as well. Multipotent mesenchymal stromal cells (MSCs) of the bone marrow are involved in immune response regulation through both intercellular interactions and secretion of various cytokines. In hematological malignancies, the bone marrow stromal microenvironment, including MSCs, is altered. Aim of this study was to describe the differences of MSCs' immunological function in the patients with acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). In ALL, malignant cells arise from the early precursor cells localized in bone marrow, while in DLBCL they arise from more differentiated B-cells. In this study, only the DLBCL patients without bone marrow involvement were included. Growth parameters, surface marker expression, genes of interest expression, and secretion pattern of bone marrow MSCs from the patients with ALL and DLBCL at the onset of the disease and in remission were studied. MSCs from the healthy donors of corresponding ages were used as controls. It has been shown that concentration of MSCs in the bone marrow of the patients with ALL is reduced at the onset of the disease and is restored upon reaching remission; in the patients with DLBCL this parameter does not change. Proliferative capacity of MSCs did not change in the patients with ALL; however, the cells of the DLBCL patients both at the onset and in remission proliferated significantly faster than those from the donors. Expression of the membrane surface markers and expression of the genes important for differentiation, immunological status maintenance, and cytokine secretion differed significantly in the MSCs of the patients from those of the healthy donors and depended on nosology of the disease. Secretomes of the MSCs varied greatly; a number of proteins associated with immune response regulation, differentiation, and maintenance of hematopoietic stem cells were depleted in the secretomes of the cells from the patients. Lymphoid neoplasia leads to dramatic changes in the functional immunological status of MSCs.
Collapse
Affiliation(s)
- Nataliya A Petinati
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| | - Aleksandra V Sadovskaya
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Natalia V Sats
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nikolai M Kapranov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Yulia O Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina A Fastova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Aminat U Magomedova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Anastasia N Vasilyeva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Olga A Aleshina
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Georgiy P Arapidi
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| | - Viktoria O Shender
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Igor P Smirnov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Olga V Pobeguts
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Nina I Drize
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Elena N Parovichnikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| |
Collapse
|
12
|
He C, Kim HI, Park J, Guo J, Huang W. The role of immune cells in different stages of atherosclerosis. Int J Med Sci 2024; 21:1129-1143. [PMID: 38774746 PMCID: PMC11103388 DOI: 10.7150/ijms.94570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/17/2024] [Indexed: 05/24/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of immune cells in the intima of arteries. Experimental and clinical evidence shows that both innate and adaptive immunity orchestrate the progression of atherosclerosis. The heterogeneous nature of immune cells within atherosclerosis lesions is important. Studies utilizing high-dimensional mass spectrometry and single-cell RNA sequencing of leukocytes from atherosclerotic lesions show the diversity and adaptability of these immune cell subtypes. Their migration, compositional changes, phenotypic alterations, and adaptive responses are key features throughout atherosclerosis progression. Understanding how these immune cells and their subtypes affect atherogenesis would help to develop novel therapeutic approaches that control atherosclerosis progression. Precise targeting of specific immune system components involved in atherosclerosis, rather than broad suppression of the immune system with anti-inflammatory agents, can more accurately regulate the progress of atherosclerosis with fewer side effects. In this review, we cover the most recent advances in the field of atherosclerosis to understand the role of various immune cells on its development. We focus on the complex network of immune cells and the interaction between the innate immune system and adaptive immune system.
Collapse
Affiliation(s)
- Cong He
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing 163319, PR China
| | - Hyo In Kim
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, United States
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Junli Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, School of Public Health, Hainan Medical University, Haikou 571199, PR China
| | - Wei Huang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199, PR China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, School of Public Health, Hainan Medical University, Haikou 571199, PR China
| |
Collapse
|
13
|
de-Apoena Reche DT, Machado NR, Fagundes BO, Bergamasco IS, de Sousa TR, do Nascimento LA, Cunha FRM, de-Oliveira MG, da-Ressureição Sgnotto F, França CN, Victor JR. IgG from Dermatophagoides pteronyssinus (Der p)-atopic individuals modulates non-atopic thymic B cell phenotype (alfa-4/beta-7) and cytokine production (IFN-γ, IL-9, and IL-10) with direct membrane interaction. Sci Rep 2024; 14:7274. [PMID: 38538762 PMCID: PMC10973508 DOI: 10.1038/s41598-024-57950-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/23/2024] [Indexed: 04/01/2024] Open
Abstract
Studies about thymic B cells are scarce in the literature, but it was suggested that they can exert modulatory and regulatory functions on the immune system. Thymic B cells can play some role in regulating the most frequent allergic background worldwide, the atopy induced by the mite Dermatophagoides pteronyssinus (Der p). Here, we aimed to evaluate if the polyclonal IgG repertoire produced by Der p-atopic individuals can influence the homing and cytokine profile of human thymic B derived from non-atopic children aged less than seven days. With this purpose, we produced polyclonal IgG formulations and cultivated human thymocytes in their presence. We also assessed IgG subclasses and the direct interaction of IgG with thymic B cell membranes. Our results could demonstrate that Der p-atopic IgG could not reduce the expression of α4β7 homing molecule as observed in response to the other IgG formulations and could reduce the frequency of IFN-γ- and IL-9-producing thymic B cells compared to the mock condition. Der p-atopic IgG could also induce thymic IL-10-producing B cells compared to control conditions. The IgG derived from Der p-atopic individuals failed to diminish the population of IL-13-producing thymic B cells, unlike the reduction observed with other IgG formulations when compared to the mock condition. All IgG formulations had similar levels of IgG subclasses and directly interacted with thymic B cell membranes. Finally, we performed experiments using peripheral non-atopic B cells where IgG effects were not observed. In conclusion, our observation demonstrates that IgG induced in allergic individuals can modulate non-atopic thymic B cells, potentially generating thymic B cells prone to allergy development, which seems to not occur in mature B cells.
Collapse
Affiliation(s)
| | - Nicolle Rakanidis Machado
- Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology, Medical School, University of Sao Paulo, Av. Dr. Enéas Carvalho de Aguiar, 500, 3rd Floor, São Paulo, SP, 05403-000, Brazil
| | - Beatriz Oliveira Fagundes
- Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology, Medical School, University of Sao Paulo, Av. Dr. Enéas Carvalho de Aguiar, 500, 3rd Floor, São Paulo, SP, 05403-000, Brazil
| | - Isabella Siuffi Bergamasco
- Post Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, SP, 04829-300, Brazil
- Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology, Medical School, University of Sao Paulo, Av. Dr. Enéas Carvalho de Aguiar, 500, 3rd Floor, São Paulo, SP, 05403-000, Brazil
| | - Thamires Rodrigues de Sousa
- Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology, Medical School, University of Sao Paulo, Av. Dr. Enéas Carvalho de Aguiar, 500, 3rd Floor, São Paulo, SP, 05403-000, Brazil
| | - Lais Alves do Nascimento
- Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology, Medical School, University of Sao Paulo, Av. Dr. Enéas Carvalho de Aguiar, 500, 3rd Floor, São Paulo, SP, 05403-000, Brazil
| | | | - Marilia Garcia de-Oliveira
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Carolina Nunes França
- Post Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, SP, 04829-300, Brazil
| | - Jefferson Russo Victor
- Post Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, SP, 04829-300, Brazil.
- Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology, Medical School, University of Sao Paulo, Av. Dr. Enéas Carvalho de Aguiar, 500, 3rd Floor, São Paulo, SP, 05403-000, Brazil.
| |
Collapse
|
14
|
Woodruff MC, Faliti CE, Sanz I. Systems biology of B cells in COVID-19. Semin Immunol 2024; 72:101875. [PMID: 38489999 DOI: 10.1016/j.smim.2024.101875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
The integration of multi-'omic datasets into complex systems-wide assessments has become a mainstay in immunologic investigation. This focus on high-dimensional data collection and analysis was on full display in the investigation of COVID-19, the respiratory illness resulting from infection by the novel coronavirus SARS-CoV-2. Particularly in the area of B cell biology, tremendous efforts in both cellular and serologic investigation have resulted in an increasingly detailed mapping of the coordinated effector, memory, and antibody secreting cell responses that underpin the development of humoral immunity in response to primary viral infection. Further, the rapid development and deployment of effective vaccines has allowed for the assessment of developing memory responses across a wide variety of immune contexts, including in patients with compromised immune function. The result has been a period of rapid gains in the understanding of B cell biology unrestricted to the study of COVID-19. Here, we outline the systems-level technologies that have been routinely implemented in these investigations throughout the pandemic, and discuss how their use has led to clear and applicable gains in pursuance of the amelioration of human infectious disease and beyond.
Collapse
Affiliation(s)
- Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| |
Collapse
|
15
|
Hu S, Song Y, Li X, Chen Q, Tang B, Chen J, Yang G, Yan H, Wang J, Wang W, Hu J, He H, Li L, Wang J. Comparative transcriptomics analysis identifies crucial genes and pathways during goose spleen development. Front Immunol 2024; 15:1327166. [PMID: 38375472 PMCID: PMC10875100 DOI: 10.3389/fimmu.2024.1327166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/17/2024] [Indexed: 02/21/2024] Open
Abstract
As the largest peripheral lymphoid organ in poultry, the spleen plays an essential role in regulating the body's immune capacity. However, compared with chickens and ducks, information about the age- and breed-related changes in the goose spleen remains scarce. In this study, we systematically analyzed and compared the age-dependent changes in the morphological, histological, and transcriptomic characteristics between Landes goose (LG; Anser anser) and Sichuan White goose (SWG; Anser cygnoides). The results showed a gradual increase in the splenic weights for both LG and SWG until week 10, while their splenic organ indexes reached the peak at week 6. Meanwhile, the splenic histological indexes of both goose breeds continuously increased with age, reaching the highest levels at week 30. The red pulp (RP) area was significantly higher in SWG than in LG at week 0, while the splenic corpuscle (AL) diameter was significantly larger in LG than in SWG at week 30. At the transcriptomic level, a total of 1710 and 1266 differentially expressed genes (DEGs) between week 0 and week 30 were identified in spleens of LG and SWG, respectively. Meanwhile, a total of 911 and 808 DEGs in spleens between LG and SWG were identified at weeks 0 and 30, respectively. Both GO and KEGG enrichment analysis showed that the age-related DEGs of LG or SWG were dominantly enriched in the Cell cycle, TGF-beta signaling, and Wnt signaling pathways, while most of the breed-related DEGs were enriched in the Neuroactive ligand-receptor interaction, Cytokine-cytokine receptor interaction, ECM-receptor interaction, and metabolic pathways. Furthermore, through construction of protein-protein interaction networks using significant DEGs, it was inferred that three hub genes including BUB1, BUB1B, and TTK could play crucial roles in regulating age-dependent goose spleen development while GRIA2, GRIA4, and RYR2 could be crucial for the breed-specific goose spleen development. These data provide novel insights into the splenic developmental differences between Chinese and European domestic geese, and the identified crucial pathways and genes are helpful for a better understanding of the mechanisms regulating goose immune functions.
Collapse
Affiliation(s)
- Shenqiang Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yang Song
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiaopeng Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Qingliang Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Bincheng Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiasen Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Guang Yang
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Haoyu Yan
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Junqi Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Wanxia Wang
- Department of Animal Production, General Station of Animal Husbandry of Sichuan Province, Chengdu, China
| | - Jiwei Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hua He
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liang Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiwen Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Livestock and Poultry Multi-Omics Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
16
|
Parola M, Pinzani M. Liver fibrosis in NAFLD/NASH: from pathophysiology towards diagnostic and therapeutic strategies. Mol Aspects Med 2024; 95:101231. [PMID: 38056058 DOI: 10.1016/j.mam.2023.101231] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
Liver fibrosis, as an excess deposition of extracellular matrix (ECM) components, results from chronic liver injury as well as persistent activation of inflammatory response and of fibrogenesis. Liver fibrosis is a major determinant for chronic liver disease (CLD) progression and in the last two decades our understanding on the major molecular and cellular mechanisms underlying the fibrogenic progression of CLD has dramatically improved, boosting pre-clinical studies and clinical trials designed to find novel therapeutic approaches. From these studies several critical concepts have emerged, starting to reveal the complexity of the pro-fibrotic microenvironment which involves very complex, dynamic and interrelated interactions between different hepatic and extrahepatic cell populations. This review will offer first a recapitulation of established and novel pathophysiological basic principles and concepts by intentionally focus the attention on NAFLD/NASH, a metabolic-related form of CLD with a high impact on the general population and emerging as a leading cause of CLD worldwide. NAFLD/NASH-related pro-inflammatory and profibrogenic mechanisms will be analysed as well as novel information on cells, mediators and signalling pathways which have taken advantage from novel methodological approaches and techniques (single cell genomics, imaging mass cytometry, novel in vitro two- and three-dimensional models, etc.). We will next offer an overview on recent advancement in diagnostic and prognostic tools, including serum biomarkers and polygenic scores, to support the analysis of liver biopsies. Finally, this review will provide an analysis of current and emerging therapies for the treatment of NAFLD/NASH patients.
Collapse
Affiliation(s)
- Maurizio Parola
- Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine - Royal Free Hospital, London, NW32PF, United Kingdom.
| |
Collapse
|
17
|
Odler B, Tieu J, Artinger K, Chen-Xu M, Arnaud L, Kitching RA, Terrier B, Thiel J, Cid MC, Rosenkranz AR, Kronbichler A, Jayne DRW. The plethora of immunomodulatory drugs: opportunities for immune-mediated kidney diseases. Nephrol Dial Transplant 2023; 38:ii19-ii28. [PMID: 37816674 DOI: 10.1093/ndt/gfad186] [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: 04/24/2023] [Indexed: 10/12/2023] Open
Abstract
In recent decades, insights into the molecular pathways involved in disease have revolutionized the treatment of autoimmune diseases. A plethora of targeted therapies have been identified and are at varying stages of clinical development in renal autoimmunity. Some of these agents, such as rituximab or avacopan, have been approved for the treatment of immune-mediated kidney disease, but kidney disease lags behind more common autoimmune disorders in new drug development. Evidence is accumulating as to the importance of adaptive immunity, including abnormalities in T-cell activation and signaling, and aberrant B-cell function. Furthermore, innate immunity, particularly the complement and myeloid systems, as well as pathologic responses in tissue repair and fibrosis, play a key role in disease. Collectively, these mechanistic studies in innate and adaptive immunity have provided new insights into mechanisms of glomerular injury in immune-mediated kidney diseases. In addition, inflammatory pathways common to several autoimmune conditions exist, suggesting that the repurposing of some existing drugs for the treatment of immune-mediated kidney diseases is a logical strategy. This new understanding challenges the clinical investigator to translate new knowledge into novel therapies leading to better disease outcomes. This review highlights promising immunomodulatory therapies tested for immune-mediated kidney diseases as a primary indication, details current clinical trials and discusses pathways that could be targeted in the future.
Collapse
Affiliation(s)
- Balazs Odler
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Johanna Tieu
- Faculty of Health and Medical Sciences, University of Adelaide; Adelaide, Australia
- Rheumatology Unit, The Queen Elizabeth Hospital, Adelaide, Australia
- Rheumatology Unit, Lyell McEwin Hospital, Adelaide, Australia
| | - Katharina Artinger
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Michael Chen-Xu
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Laurent Arnaud
- National Reference Center for Rare Auto-immune and Systemic Diseases Est Sud-Est (RESO), Strasbourg, France
| | - Richard A Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
- Departments of Nephrology and Paediatric Nephrology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Benjamin Terrier
- Department of Internal Medicine, National Reference Center for Autoimmune Diseases, Hôpital Cochin, Assistance Publique Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| | - Jens Thiel
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria C Cid
- Department of Autoimmune Diseases, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Alexander R Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Kronbichler
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria
| | - David R W Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Haas KM. Noncanonical B Cells: Characteristics of Uncharacteristic B Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1257-1265. [PMID: 37844278 PMCID: PMC10593487 DOI: 10.4049/jimmunol.2200944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/12/2023] [Indexed: 10/18/2023]
Abstract
B lymphocytes were originally described as a cell type uniquely capable of secreting Abs. The importance of T cell help in Ab production was revealed soon afterward. Following these seminal findings, investigators made great strides in delineating steps in the conventional pathway that B cells follow to produce high-affinity Abs. These studies revealed generalized, or canonical, features of B cells that include their developmental origin and paths to maturation, activation, and differentiation into Ab-producing and memory cells. However, along the way, examples of nonconventional B cell populations with unique origins, age-dependent development, tissue localization, and effector functions have been revealed. In this brief review, features of B-1a, B-1b, marginal zone, regulatory, killer, NK-like, age-associated, and atypical B cells are discussed. Emerging work on these noncanonical B cells and functions, along with the study of their significance for human health and disease, represents an exciting frontier in B cell biology.
Collapse
Affiliation(s)
- Karen M Haas
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
19
|
Satitsuksanoa P, Iwasaki S, Boersma J, Bel Imam M, Schneider SR, Chang I, van de Veen W, Akdis M. B cells: The many facets of B cells in allergic diseases. J Allergy Clin Immunol 2023; 152:567-581. [PMID: 37247640 DOI: 10.1016/j.jaci.2023.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/30/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
B cells play a key role in our immune system through their ability to produce antibodies, suppress a proinflammatory state, and contribute to central immune tolerance. We aim to provide an in-depth knowledge of the molecular biology of B cells, including their origin, developmental process, types and subsets, and functions. In allergic diseases, B cells are well known to induce and maintain immune tolerance through the production of suppressor cytokines such as IL-10. Similarly, B cells protect against viral infections such as severe acute respiratory syndrome coronavirus 2 that caused the recent coronavirus disease 2019 pandemic. Considering the unique and multifaceted functions of B cells, we hereby provide a comprehensive overview of the current knowledge of B-cell biology and its clinical applications in allergic diseases, organ transplantation, and cancer.
Collapse
Affiliation(s)
- Pattraporn Satitsuksanoa
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland.
| | - Sayuri Iwasaki
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Wageningen University & Research, Wageningen, The Netherlands
| | - Jolien Boersma
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Wageningen University & Research, Wageningen, The Netherlands
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Iris Chang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Sean N. Parker Centre for Allergy and Asthma Research, Department of Medicine, Stanford University, Palo Alto, Calif
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland.
| |
Collapse
|
20
|
Petagine L, Zariwala MG, Patel VB. Non-alcoholic fatty liver disease: Immunological mechanisms and current treatments. World J Gastroenterol 2023; 29:4831-4850. [PMID: 37701135 PMCID: PMC10494768 DOI: 10.3748/wjg.v29.i32.4831] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/14/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) causes significant global disease burden and is a leading cause of mortality. NAFLD induces a myriad of aberrant changes in hepatocytes at both the cellular and molecular level. Although the disease spectrum of NAFLD is widely recognised, the precise triggers for disease progression are still to be fully elucidated. Furthermore, the propagation to cirrhosis is poorly understood. Whilst some progress in terms of treatment options have been explored, an incomplete understanding of the hepatic cellular and molecular alterations limits their clinical utility. We have therefore reviewed some of the key pathways responsible for the pathogenesis of NAFLD such as innate and adaptative immunity, lipotoxicity and fibrogenesis, and highlighted current trials and treatment options for NAFLD patients.
Collapse
Affiliation(s)
- Lucy Petagine
- Centre for Nutraceuticals, School of Life Sciences, University of Westminster, London W1W6UW, United Kingdom
| | - Mohammed Gulrez Zariwala
- Centre for Nutraceuticals, School of Life Sciences, University of Westminster, London W1W6UW, United Kingdom
| | - Vinood B Patel
- Centre for Nutraceuticals, School of Life Sciences, University of Westminster, London W1W6UW, United Kingdom
| |
Collapse
|
21
|
Kahan R, Cray PL, Abraham N, Gao Q, Hartwig MG, Pollara JJ, Barbas AS. Sterile inflammation in liver transplantation. Front Med (Lausanne) 2023; 10:1223224. [PMID: 37636574 PMCID: PMC10449546 DOI: 10.3389/fmed.2023.1223224] [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: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Sterile inflammation is the immune response to damage-associated molecular patterns (DAMPs) released during cell death in the absence of foreign pathogens. In the setting of solid organ transplantation, ischemia-reperfusion injury results in mitochondria-mediated production of reactive oxygen and nitrogen species that are a major cause of uncontrolled cell death and release of various DAMPs from the graft tissue. When properly regulated, the immune response initiated by DAMP-sensing serves as means of damage control and is necessary for initiation of recovery pathways and re-establishment of homeostasis. In contrast, a dysregulated or overt sterile inflammatory response can inadvertently lead to further injury through recruitment of immune cells, innate immune cell activation, and sensitization of the adaptive immune system. In liver transplantation, sterile inflammation may manifest as early graft dysfunction, acute graft failure, or increased risk of immunosuppression-resistant rejection. Understanding the mechanisms of the development of sterile inflammation in the setting of liver transplantation is crucial for finding reliable biomarkers that predict graft function, and for development of therapeutic approaches to improve long-term transplant outcomes. Here, we discuss the recent advances that have been made to elucidate the early signs of sterile inflammation and extent of damage from it. We also discuss new therapeutics that may be effective in quelling the detrimental effects of sterile inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrew S. Barbas
- Duke Ex-Vivo Organ Lab (DEVOL)—Division of Abdominal Transplant Surgery, Duke University, Durham, NC, United States
| |
Collapse
|
22
|
Castleman MJ, Santos AL, Lesteberg KE, Maloney JP, Janssen WJ, Mould KJ, Beckham JD, Pelanda R, Torres RM. Activation and pro-inflammatory cytokine production by unswitched memory B cells during SARS-CoV-2 infection. Front Immunol 2023; 14:1213344. [PMID: 37638016 PMCID: PMC10449608 DOI: 10.3389/fimmu.2023.1213344] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Memory B cells are comprised of unswitched (CD27+IgD+) and switched (CD27+IgD-) subsets. The origin and function of unswitched human memory B cells are debated in the literature, whereas switched memory B cells are primed to respond to recurrent infection. Unswitched memory B cells have been described to be reduced in frequency with severe SARS-CoV2 infection and here we characterize their activation status, BCR functionality, and contribution to virally-induced cytokine production. Analyses of whole blood from healthy individuals, people immunized against SARS-CoV2, and those who have had mild and severe SARS-CoV2 infection, confirm a reduction in the frequency of unswitched memory B cells during severe SARS-CoV2 infection and demonstrate this reduction is associated with increased levels of systemic TNFα. We further document how severe viral infection is associated with an increased frequency of 'IgD+' only memory B cells that correlate with increased IgG autoantibody levels. Unswitched and switched memory B cells from severe SARS-CoV2 infection displayed evidence of heightened activation with a concomitant reduction in the expression of the inhibitory receptor CD72. Functionally, both populations of memory B cells from severe SARS-COV2 infection harbored a signaling-competent BCR that displayed enhanced BCR signaling activity in the unswitched population. Finally, we demonstrate that B cells from mild SARS-CoV2 infection are poised to secrete pro-inflammatory cytokines IL-6 and TNFα. Importantly, unswitched memory B cells were a major producer of IL-6 and switched memory B cells were a major producer of TNFα in response to viral TLR ligands. Together these data indicate that B cells contribute to the inflammatory milieu during viral infection.
Collapse
Affiliation(s)
- Moriah J. Castleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Adriana Luna Santos
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kelsey E. Lesteberg
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO, United States
| | - James P. Maloney
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - William J. Janssen
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Kara J. Mould
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - J. David Beckham
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional VA, Medical Center, Aurora, CO, United States
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
23
|
Popa-Fotea NM, Ferdoschi CE, Micheu MM. Molecular and cellular mechanisms of inflammation in atherosclerosis. Front Cardiovasc Med 2023; 10:1200341. [PMID: 37600028 PMCID: PMC10434786 DOI: 10.3389/fcvm.2023.1200341] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Atherosclerosis and its complications are a major cause of morbidity and mortality worldwide in spite of the improved medical and invasive treatment in terms of revascularization. Atherosclerosis is a dynamic, multi-step process in which inflammation is a ubiquitous component participating in the initiation, development, and entanglements of the atherosclerotic plaque. After activation, the immune system, either native or acquired, is part of the atherosclerotic dynamics enhancing the pro-atherogenic function of immune or non-immune cells, such as endothelial cells, smooth muscle cells, or platelets, through mediators such as cytokines or directly by cell-to-cell interaction. Cytokines are molecules secreted by the activated cells mentioned above that mediate the inflammatory component of atherosclerosis whose function is to stimulate the immune cells and the production of further cytokines. This review provides insights of the cell axis activation and specific mechanisms and pathways through which inflammation actuates atherosclerosis.
Collapse
Affiliation(s)
- Nicoleta-Monica Popa-Fotea
- Department 4 Cardio-Thoracic Pathology, University of Medicine and Pharmacy “Carol Davila,”Bucharest, Romania
- Cardiology Department, Emergency Clinical Hospital, Bucharest, Romania
| | - Corina-Elena Ferdoschi
- Department 4 Cardio-Thoracic Pathology, University of Medicine and Pharmacy “Carol Davila,”Bucharest, Romania
| | | |
Collapse
|
24
|
Danielsen AK, Damgaard C, Massarenti L, Østrup P, Riis Hansen P, Holmstrup P, Nielsen CH. B-cell cytokine responses to Porphyromonas gingivalis in patients with periodontitis and healthy controls. J Periodontol 2023; 94:997-1007. [PMID: 36715211 DOI: 10.1002/jper.22-0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/24/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cytokine-producing B cells play a well-established role in modifying immune responses in chronic inflammatory diseases. We characterized B-cell cytokine responses against periodontitis-associated bacteria in patients with periodontitis. METHODS Blood and saliva samples were collected from patients with periodontitis grade B (N = 31) or grade C (N = 25), and 25 healthy controls (HCs). Mononuclear cells were stimulated with Porphyromonas gingivalis, Fusobacterium nucleatum, Staphylococcus epidermidis, or Cutibacterium acnes, and B-cell production of tumor necrosis factor (TNF)-α, interleukin (IL)-6, interferon (IFN)-γ, IL-10 and transforming growth factor (TGF)-β by B cells was assessed by flow cytometry. RESULTS HCs had higher baseline frequencies of B cells producing IFN-γ or TNF-α than grade B patients, but only B cells from grade B patients showed significant differentiation into IFN-γ-, TNF-α-, TGF-β-, or IL-10-producing cells after challenge with P. gingivalis and into IFN-γ-, TGF-β-, or IL-10-producing cells after challenge F. nucleatum. Notably, the baseline frequency of IL-10-producing B cells from grade C patients correlated inversely with clinical attachment loss (AL). The major proportion of the IFN-γ- and TGF-β-producing B cells were CD27+ memory cells, while the IL-10-producing B cells were mainly CD27- CD5- . CONCLUSIONS B cells from grade B patients, particularly those harboring P. gingivalis, showed proinflammatory B-cell responses to P. gingivalis. Moreover, the baseline frequency of IL-10-producing B cells in the grade C group correlated inversely with AL, suggesting a diminished immunoregulatory capacity of IL-10-producing B cells in these patients.
Collapse
Affiliation(s)
- Anne Katrine Danielsen
- Faculty of Health and Medical Sciences, Section for Oral, Biology and Immunopathology, Department of Odontology, University of Copenhagen, Copenhagen, Denmark
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Damgaard
- Faculty of Health and Medical Sciences, Section for Oral, Biology and Immunopathology, Department of Odontology, University of Copenhagen, Copenhagen, Denmark
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Laura Massarenti
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter Østrup
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Riis Hansen
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Palle Holmstrup
- Faculty of Health and Medical Sciences, Section for Oral, Biology and Immunopathology, Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| | - Claus H Nielsen
- Faculty of Health and Medical Sciences, Section for Oral, Biology and Immunopathology, Department of Odontology, University of Copenhagen, Copenhagen, Denmark
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
25
|
Sanami S, Nazarian S, Ahmad S, Raeisi E, Tahir Ul Qamar M, Tahmasebian S, Pazoki-Toroudi H, Fazeli M, Ghatreh Samani M. In silico design and immunoinformatics analysis of a universal multi-epitope vaccine against monkeypox virus. PLoS One 2023; 18:e0286224. [PMID: 37220125 DOI: 10.1371/journal.pone.0286224] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Monkeypox virus (MPXV) outbreaks have been reported in various countries worldwide; however, there is no specific vaccine against MPXV. In this study, therefore, we employed computational approaches to design a multi-epitope vaccine against MPXV. Initially, cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL), linear B lymphocytes (LBL) epitopes were predicted from the cell surface-binding protein and envelope protein A28 homolog, both of which play essential roles in MPXV pathogenesis. All of the predicted epitopes were evaluated using key parameters. A total of 7 CTL, 4 HTL, and 5 LBL epitopes were chosen and combined with appropriate linkers and adjuvant to construct a multi-epitope vaccine. The CTL and HTL epitopes of the vaccine construct cover 95.57% of the worldwide population. The designed vaccine construct was found to be highly antigenic, non-allergenic, soluble, and to have acceptable physicochemical properties. The 3D structure of the vaccine and its potential interaction with Toll-Like receptor-4 (TLR4) were predicted. Molecular dynamics (MD) simulation confirmed the vaccine's high stability in complex with TLR4. Finally, codon adaptation and in silico cloning confirmed the high expression rate of the vaccine constructs in strain K12 of Escherichia coli (E. coli). These findings are very encouraging; however, in vitro and animal studies are needed to ensure the potency and safety of this vaccine candidate.
Collapse
Affiliation(s)
- Samira Sanami
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahin Nazarian
- Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, CA, United States of America
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Elham Raeisi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Muhammad Tahir Ul Qamar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Shahram Tahmasebian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hamidreza Pazoki-Toroudi
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Fazeli
- WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Ghatreh Samani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
26
|
Lu L, Ma W, Johnson CH, Khan SA, Irwin ML, Pusztai L. In silico designed mRNA vaccines targeting CA-125 neoantigen in breast and ovarian cancer. Vaccine 2023; 41:2073-2083. [PMID: 36813666 PMCID: PMC10064809 DOI: 10.1016/j.vaccine.2023.02.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023]
Abstract
Somatic mutation-derived neoantigens are associated with patient survival in breast and ovarian cancer. These neoantigens are targets for cancer, as shown by the implementation of neoepitope peptides as cancer vaccines. The success of cost-effective multi-epitope mRNA vaccines against SARS-Cov-2 in the pandemic established a model for reverse vaccinology. In this study, we aimed to develop an in silico pipeline designing an mRNA vaccine of the CA-125 neoantigen against breast and ovarian cancer, respectively. Using immuno-bioinformatics tools, we predicted cytotoxic CD8+ T cell epitopes based on somatic mutation-driven neoantigens of CA-125 in breast or ovarian cancer, constructed a self-adjuvant mRNA vaccine with CD40L and MHC-I -targeting domain to enhance cross-presentation of neoepitopes by dendritic cells. With an in silico ImmSim algorithm, we estimated the immune responses post-immunization, showing IFN-γ and CD8+ T cell response. The strategy described in this study may be scaled up and implemented to design precision multi-epitope mRNA vaccines by targeting multiple neoantigens.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06510, USA; Yale Cancer Center, Yale University, New Haven, CT 06510, USA.
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center and Sanford Stem Cell Clinical Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Caroline H Johnson
- Yale Cancer Center, Yale University, New Haven, CT 06510, USA; Department of Environmental Health Science, Yale School of Public Health, Yale University, New Haven, CT 06510, USA
| | - Sajid A Khan
- Yale Cancer Center, Yale University, New Haven, CT 06510, USA; Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Melinda L Irwin
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06510, USA; Yale Cancer Center, Yale University, New Haven, CT 06510, USA
| | - Lajos Pusztai
- Yale Cancer Center, Yale University, New Haven, CT 06510, USA; Department of Medical Oncology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
27
|
B cells promote granulomatous inflammation during chronic Mycobacterium tuberculosis infection in mice. PLoS Pathog 2023; 19:e1011187. [PMID: 36888692 PMCID: PMC9994760 DOI: 10.1371/journal.ppat.1011187] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/05/2023] [Indexed: 03/09/2023] Open
Abstract
The current study reveals that in chronic TB, the B cell-deficient μMT strain, relative to wild-type (WT) C57BL/6 mice, displays in the lungs lower levels of inflammation that are associated with decreased CD4+ T cell proliferation, diminished Th1 response, and enhanced levels of interleukin (IL)-10. The latter result raises the possibility that B cells may restrict lung expression of IL-10 in chronic TB. These observations are recapitulated in WT mice depleted for B cells using anti-CD20 antibodies. IL-10 receptor (IL-10R) blockade reverses the phenotypes of decreased inflammation and attenuated CD4+ T cell responses in B cell-depleted mice. Together, these results suggest that in chronic murine TB, B cells, by virtue of their capacity to restrict expression of the anti-inflammatory and immunosuppressive IL-10 in the lungs, promote the development of a robust protective Th1 response, thereby optimizing anti-TB immunity. This vigorous Th1 immunity and restricted IL-10 expression may, however, allow the development of inflammation to a level that can be detrimental to the host. Indeed, decreased lung inflammation observed in chronically infected B cell-deficient mice, which exhibit augmented lung IL-10 levels, is associated with a survival advantage relative to WT animals. Collectively, the results reveal that in chronic murine TB, B cells play a role in modulating the protective Th1 immunity and the anti-inflammatory IL-10 response, which results in augmentation of lung inflammation that can be host-detrimental. Intriguingly, in tuberculous human lungs, conspicuous B cell aggregates are present in close proximity to tissue-damaging lesions manifesting necrosis and cavitation, suggesting the possibility that in human TB, B cells may contribute to the development of exacerbated pathology that is known to promote transmission. Since transmission is a major hindrance to TB control, investigating into whether B cells can shape the development of severe pulmonic pathological responses in tuberculous individuals is warranted.
Collapse
|
28
|
Sayin I, Chong AS. Beyond Adaptive Alloreactivity: Contribution of Innate B Cells to Allograft Inflammation and Rejection. Transplantation 2023; 107:98-104. [PMID: 36404414 PMCID: PMC9772142 DOI: 10.1097/tp.0000000000004377] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Innate B cells are a heterogeneous group of cells that function in maintaining homeostatic levels of circulating natural antibodies and being the first line of defense against infections. Innate B-1 cells and marginal zone B cells may relocate to lymphoid follicles and differentiate into cytokine and antibody-secreting cells in T-independent and T-dependent manners. Although marginal zone B cells are widely described in humans, the presence of B-1 cells is more controversial. Here, we review the basic features of the innate B-cell subsets identified in mice and their equivalent in humans, as well as their potential roles in transplantation. We summarize the findings of Cascalho and colleagues on the unexpected protective role of tumor necrosis factor receptor superfamily member 13B in regulating circulating levels of protective natural immunoglobulin M, and the studies by Zorn and colleagues on the potential pathogenic role for polyreactive innate B cells infiltrating allograft explants. Finally, we discuss our studies that took a transcriptomic approach to identify innate B cells infiltrating kidney allografts with antibody-mediated rejection and to demonstrate that local antigens within the allograft together with inflammation may induce a loss of B-cell tolerance.
Collapse
Affiliation(s)
- Ismail Sayin
- Department of Surgery, The University of Chicago, Chicago, Illinois, United States
| | - Anita S. Chong
- Department of Surgery, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
29
|
Li K, Romero M, Cañardo M, Garcia D, Diaz A, Blomberg BB, Frasca D. B cells from old mice induce the generation of inflammatory T cells through metabolic pathways. Mech Ageing Dev 2023; 209:111742. [PMID: 36309082 DOI: 10.1016/j.mad.2022.111742] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
We have measured the capacity of B cells from young and old mice to induce the differentiation of naïve CD4 + T cells from young mice into pro-inflammatory subsets. We found that only B cells from old mice are inflammatory and induce in vitro secretion of the pro-inflammatory cytokines IL-17A and IFN-γ by T cells. In co-culture experiments, B cells from old mice showed a strong helper function on T cells from young mice, making them pro-inflammatory, and this effect is regulated by metabolic pathways, mainly anaerobic glycolysis, leading to increased RNA expression of the enzyme lactate dehydrogenase (LDHA) and increased secretion of lactate. These results have indicated that lactate is a crucial player of the B cell-induced polarization of T cells. When we measured the effects of lactate on isolated CD4 + T cells from young mice, we found that lactate increases RNA expression of LDHA, secretion of pro-inflammatory cytokines and NF-kB activation. Moreover, lactate effects in culture can be abrogated in the presence of the specific inhibitor of LDHA, FX11. These results altogether may have relevant clinical implications and suggest novel targets for therapeutic interventions in patients with inflammatory conditions and diseases.
Collapse
Affiliation(s)
- Kevin Li
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Maria Romero
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Macarena Cañardo
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Denisse Garcia
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Alain Diaz
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA; Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL USA
| | - Daniela Frasca
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA; Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL USA.
| |
Collapse
|
30
|
Oleinika K, Slisere B, Catalán D, Rosser EC. B cell contribution to immunometabolic dysfunction and impaired immune responses in obesity. Clin Exp Immunol 2022; 210:263-272. [PMID: 35960996 PMCID: PMC9384752 DOI: 10.1093/cei/uxac079] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/15/2022] [Accepted: 08/09/2022] [Indexed: 11/14/2022] Open
Abstract
Obesity increases the risk of type 2 diabetes mellitus, cardiovascular disease, fatty liver disease, and cancer. It is also linked with more severe complications from infections, including COVID-19, and poor vaccine responses. Chronic, low-grade inflammation and associated immune perturbations play an important role in determining morbidity in people living with obesity. The contribution of B cells to immune dysregulation and meta-inflammation associated with obesity has been documented by studies over the past decade. With a focus on human studies, here we consolidate the observations demonstrating that there is altered B cell subset composition, differentiation, and function both systemically and in the adipose tissue of individuals living with obesity. Finally, we discuss the potential factors that drive B cell dysfunction in obesity and propose a model by which altered B cell subset composition in obesity underlies dysfunctional B cell responses to novel pathogens.
Collapse
Affiliation(s)
- Kristine Oleinika
- Correspondence: Kristine Oleinika, Department of Internal Diseases, Riga Stradins University, Riga, Latvia.
| | - Baiba Slisere
- Department of Doctoral Studies, Riga Stradins University, Riga, Latvia
- Joint Laboratory, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Diego Catalán
- Programa Disciplinario de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH and Department of Rheumatology, Division of Medicine, University College London, London, UK
| |
Collapse
|
31
|
Conde L, Maciel G, de Assis GM, Freire-de-Lima L, Nico D, Vale A, Freire-de-Lima CG, Morrot A. Humoral response in Leishmaniasis. Front Cell Infect Microbiol 2022; 12:1063291. [PMID: 36579347 PMCID: PMC9791258 DOI: 10.3389/fcimb.2022.1063291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022] Open
Abstract
Leishmaniasis presents different types of clinical manifestations that can be divided into cutaneous leishmaniasis and visceral leishmaniasis. The host's immune system, associated with genetic and nutritional factors, is strongly involved in the evolution of the disease or parasite escape. Humoral immunity is characterized by the production of antibodies capable of promoting neutralization, opsonization, and activation of the complement system. In this scenario, B lymphocytes produce antibodies that play an important role in Leishmania infection although neglected for a long time. Thus, relevant aspects in the establishment of Leishmania infection will be addressed, highlighting the importance of humoral immunity during the entire process of Leishmania infection.
Collapse
Affiliation(s)
- Luciana Conde
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriela Maciel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Meira de Assis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dirlei Nico
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Vale
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Alexandre Morrot
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil,*Correspondence: Alexandre Morrot,
| |
Collapse
|
32
|
Xiong H, Tang Z, Xu Y, Shi Z, Guo Z, Liu X, Tan G, Ai X, Guo Q. CD19 +CD24 highCD27 + B cell and interleukin 35 as potential biomarkers of disease activity in systemic lupus erythematosus patients. Adv Rheumatol 2022; 62:48. [PMID: 36494762 DOI: 10.1186/s42358-022-00279-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/19/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that associates with aberrant activation of B lymphocytes and excessive autoantibodies. Interleukin 10 (IL-10)/interleukin 35 (IL-35) and IL-10/IL-35-producing regulatory B cells have been demonstrated to possess immunosuppressive functions during systemic lupus erythematosus. Here, we detected the proportion of CD19+CD24highCD27+ B cells as well as IL-10 and IL-35 levels in peripheral blood of SLE patients and healthy individuals, and investigated their relations with clinical features of SLE. METHODS 41 SLE patients and 25 healthy controls were recruited. The patients were divided into groups based on SLEDAI score, anti-dsDNA antibody, rash, nephritis and hematological disorder. Flow cytometry was used to detect the proportion of CD24hiCD27+ B cells. ELISA was used to detect serum levels of IL-10 and IL-35. RESULTS Our results showed that the CD19+CD24highCD27+ B population was decreased in active SLE patients, and anti-correlated with the disease activity. Of note, we found significant increase of IL-10 and decrease of IL-35 in SLE patients with disease activity score > 4, lupus nephritis or hematological disorders compared to those without related clinical features. CONCLUSIONS Reduced CD19+CD24highCD27+ B cells expression may be involved in the pathogenesis of SLE. Moreover, we supposed that IL-35 instead of IL-10 played a crucial role in immune regulation during SLE disease.
Collapse
Affiliation(s)
- Hui Xiong
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetic and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zengqi Tang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Ying Xu
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zhixuan Guo
- Department of Dermatology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518040, Guangdong, China
| | - Xiuting Liu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Guozhen Tan
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Xuechen Ai
- Department of Dermatology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, Guangdong, China.
| | - Qing Guo
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
33
|
Immune cell interactions in tuberculosis. Cell 2022; 185:4682-4702. [PMID: 36493751 DOI: 10.1016/j.cell.2022.10.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 12/13/2022]
Abstract
Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.
Collapse
|
34
|
Stabel JR, Bannantine JP, Humphrey S. B cell phenotypes and maturation states in cows naturally infected with Mycobacterium avium subsp. Paratuberculosis. PLoS One 2022; 17:e0278313. [PMID: 36477266 PMCID: PMC9728927 DOI: 10.1371/journal.pone.0278313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Little is known about the role that B cells play in immune responses to infection with the intracellular pathogen, Mycobacterium avium subsp. paratuberculosis (MAP). Traditionally, the role of B cells has been constrained to their function as antibody-producing cells, however, antibodies are not thought to play a protective role in mycobacterial infections. The present study was designed to characterize B cell subpopulations as well as activation/maturation states in cattle with paratuberculosis. Peripheral blood mononuclear cells (PBMCs) were isolated from noninfected control cows (n = 8); as well cattle naturally infected with MAP in the subclinical (n = 8) and clinical (n = 7) stage of infection and stimulated with MAP antigen for 6 days. MAP infection resulted in greater numbers of total B cells for clinical cows compared to control noninfected cows. The major subpopulation in freshly isolated PBMCs in clinical cows was B-1a B cells, but this shifted to a composite of both B-1a and B-2 B cells upon stimulation of PBMCs with either MAP antigen or pokeweed mitogen, with higher numbers of B-2 B cells. Early B cells were observed to predominate the population of B cells in PBMCs, with lesser populations of germinal B cells, memory B cells and plasma cells. These subpopulations were elevated in clinical cows upon stimulation of PBMCs with MAP antigen, except for plasma cells which were lower compared to control noninfected cows. Increased numbers of B cells in clinical cows aligned with higher expression of B cell markers such as MAPK1/3, BTG1, Bcl2, CD79A and SWAP70, depending upon in vitro stimulation with either mitogen or antigen. This would indicate that the B cells were capable of activation but were anti-apoptotic in nature. The shift to B-2 B cells in the periphery of clinical cows seems to be indicative of an expansion of memory B cells, rather than plasma cells. This may be a last attempt by the host to control the rampant inflammatory state associated with advanced clinical disease.
Collapse
Affiliation(s)
- J. R. Stabel
- Infectious Bacterial Diseases of Livestock Research Unit, USDA-ARS, National Animal Disease Center, Ames, IA, United States of America
- * E-mail:
| | - J. P. Bannantine
- Infectious Bacterial Diseases of Livestock Research Unit, USDA-ARS, National Animal Disease Center, Ames, IA, United States of America
| | - S. Humphrey
- Microscopy Services Department, USDA-ARS, National Animal Disease Center, Ames, IA, United States of America
| |
Collapse
|
35
|
Zhang P, Zhu H. Cytokines in Thyroid-Associated Ophthalmopathy. J Immunol Res 2022; 2022:2528046. [PMID: 36419958 PMCID: PMC9678454 DOI: 10.1155/2022/2528046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 09/07/2023] Open
Abstract
Thyroid-associated ophthalmopathy (TAO), also known as thyroid eye disease (TED) or Graves' orbitopathy (GO), is a complex autoimmune condition causing visual impairment, disfigurement, and harm to patients' physical and mental health. The pathogenesis of TAO has not been fully elucidated, and the mainstream view is that coantigens shared by the thyroid and orbit trigger remodeling of extraocular muscles and orbital connective tissues through an inflammatory response. In recent years, cytokines and the immune responses they mediate have been crucial in disease progression, and currently, common evidence has shown that drugs targeting cytokines, such as tocilizumab, infliximab, and adalimumab, may be novel targets for therapy. In this review, we summarize the research development of different cytokines in TAO pathogenesis in the hope of discovering new therapeutic targets.
Collapse
Affiliation(s)
- Pengbo Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huang Zhu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
36
|
David K, Friedlander G, Pellegrino B, Radomir L, Lewinsky H, Leng L, Bucala R, Becker-Herman S, Shachar I. CD74 as a regulator of transcription in normal B cells. Cell Rep 2022; 41:111572. [DOI: 10.1016/j.celrep.2022.111572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/07/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
|
37
|
Gavitt TD, Mara AB, Goodridge ML, Ozyck RG, Reinhardt E, Miller JM, Hunte M, Tulman ER, Frasca Jr S, Silbart LK, Geary SJ, Szczepanek SM. B cells oppose Mycoplasma pneumoniae vaccine enhanced disease and limit bacterial colonization of the lungs. NPJ Vaccines 2022; 7:130. [PMID: 36310317 PMCID: PMC9618410 DOI: 10.1038/s41541-022-00556-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Development of an effective vaccine for Mycoplasma pneumoniae has been hindered by reports of Vaccine Enhanced Disease (VED) in test subjects vaccinated and challenged in studies conducted in the 1960s. The exact mechanism of disease exacerbation has yet to be fully described, but host immune responses to Lipid-Associated Membrane Proteins (LAMPs) lipoprotein lipid moieties have been implicated. LAMPs-induced exacerbation appears to involve helper T cell recall responses, due in part to their influence on neutrophil recruitment and subsequent inflammatory responses in the lung. Herein, we characterized the functions of host B cell responses to M. pneumoniae LAMPs and delipidated-LAMPs (dLAMPs) by conducting passive transfer and B cell depletion studies to assess their contribution to disease exacerbation or protection using a BALB/c mouse model. We found that antibody responses to M. pneumoniae LAMPs and dLAMPs differ in magnitude, but not in isotype or subclass. Passive transfer, dLAMP denaturation, and monoclonal antibody studies indicate that antibodies do not cause VED, but do appear to contribute to control of bacterial loads in the lungs. Depletion of B cells prior to LAMPs-vaccination results in significantly enhanced pathology in comparison to B cell competent controls, suggesting a possible regulatory role of B cells distinct from antibody secretion. Taken together, our findings suggest that B cell antibody responses to M. pneumoniae contribute to, but are insufficient for protection against challenge on their own, and that other functional properties of B cells are necessary to limit exacerbation of disease in LAMPs-vaccinated mice after infection.
Collapse
Affiliation(s)
- Tyler D. Gavitt
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Arlind B. Mara
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Meagan L. Goodridge
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA
| | - Rosemary Grace Ozyck
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA
| | - Emily Reinhardt
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,Connecticut Veterinary Medical Diagnostic Laboratory, Storrs, CT 06238 USA
| | - Jeremy M. Miller
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Morgan Hunte
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,grid.417555.70000 0000 8814 392XPresent Address: Sanofi, Meriden, CT 06450 USA
| | - Edan R. Tulman
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Salvatore Frasca Jr
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA
| | - Lawrence K. Silbart
- grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Department of Allied of Health Sciences, University of Connecticut, Storrs, CT 06238 USA
| | - Steven J. Geary
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| | - Steven M. Szczepanek
- grid.63054.340000 0001 0860 4915Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06238 USA ,grid.63054.340000 0001 0860 4915Center of Excellence for Vaccine Research, University of Connecticut, Storrs, CT 06238 USA ,US Animal Vaccinology Research Coordination Network, Storrs, CT 06238 USA
| |
Collapse
|
38
|
Interplay between fat cells and immune cells in bone: Impact on malignant progression and therapeutic response. Pharmacol Ther 2022; 238:108274. [DOI: 10.1016/j.pharmthera.2022.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022]
|
39
|
Cherukuri A, Rothstein DM. Regulatory and transitional B cells: potential biomarkers and therapeutic targets in organ transplantation. Curr Opin Organ Transplant 2022; 27:385-391. [PMID: 35950881 PMCID: PMC9474638 DOI: 10.1097/mot.0000000000001010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF THE REVIEW Regulatory B cells (Bregs) play a prominent role in various disease settings. While progress has been hindered by the lack of a specific Breg marker, new findings highlight their role modulating the alloimmune response and promoting allograft survival. RECENT FINDINGS Herein, we focus on the recent advances in Breg biology and their role in transplantation. We review studies showing that T-cell immunoglobulin and mucin domain 1 (TIM-1) is an inclusive and functional Breg marker in mice that may have human relevance. We highlight the utility of the B cell interleukin-10/tumor necrosis factor-alpha (IL-10/TNFα) ratio in identifying underlying immunological reactivity and predicting clinical outcomes in kidney transplantation. This may identify patients requiring more immunosuppression and provide insight into potential therapeutic approaches that can modulate the Breg: B effector cell (Beff) balance. SUMMARY Emerging data support Bregs as potent modulators of immune responses in humans. Their ability to promote allograft survival must await development of approaches to expand Bregs in vitro/in vivo . The low IL-10/TNFα ratio reflecting decreased Breg/Beff balance, predicts acute rejection (AR) and poorer outcomes in renal transplantation. It remains to be determined whether this paradigm can be extended to other allografts and whether therapy aiming to correct the relative deficiency of Bregs will improve outcomes.
Collapse
Affiliation(s)
- Aravind Cherukuri
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, PA, USA
| | - David M. Rothstein
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, PA, USA
| |
Collapse
|
40
|
Chen TX, Fan YT, Peng BW. Distinct mechanisms underlying therapeutic potentials of CD20 in neurological and neuromuscular disease. Pharmacol Ther 2022; 238:108180. [DOI: 10.1016/j.pharmthera.2022.108180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
41
|
Vaccinomics to Design a Multiepitope Vaccine against Legionella pneumophila. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4975721. [PMID: 36164443 PMCID: PMC9509222 DOI: 10.1155/2022/4975721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022]
Abstract
Legionella pneumophila is found in the natural aquatic environment and can resist a wide range of environmental conditions. There are around fifty species of Legionella, at least twenty-four of which are directly linked to infections in humans. L. pneumophila is the cause of Legionnaires' disease, a potentially lethal form of pneumonia. By blocking phagosome-lysosome fusion, L. pneumophila lives and proliferates inside macrophages. For this disease, there is presently no authorized multiepitope vaccine available. For the multi-epitope-based vaccine (MEBV), the best antigenic candidates were identified using immunoinformatics and subtractive proteomic techniques. Several immunoinformatics methods were utilized to predict B and T cell epitopes from vaccine candidate proteins. To construct an in silico vaccine, epitopes (07 CTL, 03 HTL, and 07 LBL) were carefully selected and docked with MHC molecules (MHC-I and MHC-II) and human TLR4 molecules. To increase the immunological response, the vaccine was combined with a 50S ribosomal adjuvant. To maximize vaccine protein expression, MEBV was cloned and reverse-translated in Escherichia coli. To prove the MEBV's efficacy, more experimental validation is required. After its development, the resulting vaccine is greatly hoped to aid in the prevention of L. pneumophila infections.
Collapse
|
42
|
Yoshizaki A, Fukasawa T, Ebata S, Yoshizaki-Ogawa A, Sato S. Involvement of B cells in the development of systemic sclerosis. Front Immunol 2022; 13:938785. [PMID: 35967355 PMCID: PMC9365989 DOI: 10.3389/fimmu.2022.938785] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare intractable systemic disease that causes fibrosis and vasculopathy against a background of autoimmune abnormalities. Although the etiology is not yet fully understood, the type of autoantibodies detected in SSc is closely associated with disease severity and prognosis, supporting that those autoimmune abnormalities play an important role in the pathogenesis of SSc. Although the direct pathogenicity of autoantibodies found in SSc is unknown, many previous studies have shown that B cells are involved in the development of SSc through a variety of functions. Furthermore, a number of clinical studies have been conducted in which B-cell depletion therapy has been tried for SSc, and many of these studies have found B-cell depletion therapy to be effective for SSc. However, the involvement of B cells in pathogenesis is complex, as they not only promote inflammation but also play an inhibitory role. This article outlines the role of B cells in the development of SSc, including the latest research.
Collapse
|
43
|
Ruiz-Ranz M, Lequerica-Fernández P, Rodríguez-Santamarta T, Suárez-Sánchez FJ, López-Pintor RM, García-Pedrero JM, de Vicente JC. Prognostic implications of preoperative systemic inflammatory markers in oral squamous cell carcinoma, and correlations with the local immune tumor microenvironment. Front Immunol 2022; 13:941351. [PMID: 35958590 PMCID: PMC9360320 DOI: 10.3389/fimmu.2022.941351] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/19/2022] Open
Abstract
Purpose The aim of this study was to investigate the prognostic significance of preoperative inflammatory markers in peripheral blood of patients with oral squamous cell carcinoma (OSCC), and to establish correlations with the infiltrate of macrophages and lymphocytes in the local immune tumor microenvironment (TME). Materials and Methods Neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), lymphocyte-monocyte ratio (LMR), and systemic immune-inflammation index (SII) were retrospectively evaluated in a cohort of 348 OSCC patients, and correlated with overall (OS) and disease-specific survival (DSS). Immunohistochemical analysis of tumoral and stromal infiltration of CD8+, CD4+, FOXP3+ and CD20+ lymphocytes and CD68+ and CD163+ macrophages was performed in a subset of 119 OSCC patient samples, and correlations further assessed. Results NLR, SII, and LMR were significantly associated with a poorer OS in univariate analysis; however, only NLR remained a significant independent predictor in the multivariate analysis (HR = 1.626, p = 0.04). NLR and SII were inversely and significantly correlated with stromal infiltration of CD8+, CD4+, and CD20+ lymphocytes. Moreover, a significant correlation between LMR was also found to significantly associate with stromal infiltration of CD8+, CD4+, and CD20+ lymphocytes, stromal CD68+ and CD163+ macrophages, and also tumoral infiltration of CD4+ and CD20+ lymphocytes. Conclusions Preoperative NLR, SII, and LMR may serve as valuable systemic markers to predict OSCC patient survival, with NLR emerging as an independent predictor of poor OS. Moreover, strong significant correlations were exclusively observed between systemic inflammatory markers and the local stromal infiltration of lymphocytes in the TME.
Collapse
Affiliation(s)
- Marta Ruiz-Ranz
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Paloma Lequerica-Fernández
- Department of Biochemistry, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Tania Rodríguez-Santamarta
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | | | - Rosa M. López-Pintor
- ORALMED Research Group, Department of Dental Clinical Specialties, School of Dentistry, Complutense University, Madrid, Spain
| | - Juana M. García-Pedrero
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Department of Otolaryngology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Juan C. de Vicente, ; Juana M. García-Pedrero,
| | - Juan C. de Vicente
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Department of Surgery, University of Oviedo, Oviedo, Spain
- *Correspondence: Juan C. de Vicente, ; Juana M. García-Pedrero,
| |
Collapse
|
44
|
Radovani B, Gudelj I. N-Glycosylation and Inflammation; the Not-So-Sweet Relation. Front Immunol 2022; 13:893365. [PMID: 35833138 PMCID: PMC9272703 DOI: 10.3389/fimmu.2022.893365] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammation is the main feature of many long-term inflammatory diseases such as autoimmune diseases, metabolic disorders, and cancer. There is a growing number of studies in which alterations of N-glycosylation have been observed in many pathophysiological conditions, yet studies of the underlying mechanisms that precede N-glycome changes are still sparse. Proinflammatory cytokines have been shown to alter the substrate synthesis pathways as well as the expression of glycosyltransferases required for the biosynthesis of N-glycans. The resulting N-glycosylation changes can further contribute to disease pathogenesis through modulation of various aspects of immune cell processes, including those relevant to pathogen recognition and fine-tuning the inflammatory response. This review summarizes our current knowledge of inflammation-induced N-glycosylation changes, with a particular focus on specific subsets of immune cells of innate and adaptive immunity and how these changes affect their effector functions, cell interactions, and signal transduction.
Collapse
Affiliation(s)
- Barbara Radovani
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Ivan Gudelj
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| |
Collapse
|
45
|
Courey-Ghaouzi AD, Kleberg L, Sundling C. Alternative B Cell Differentiation During Infection and Inflammation. Front Immunol 2022; 13:908034. [PMID: 35812395 PMCID: PMC9263372 DOI: 10.3389/fimmu.2022.908034] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023] Open
Abstract
Long-term protective immunity to infectious disease depends on cell-mediated and humoral immune responses. Induction of a strong humoral response relies on efficient B cell activation and differentiation to long-lived plasma cells and memory B cells. For many viral or bacterial infections, a single encounter is sufficient to induce such responses. In malaria, the induction of long-term immunity can take years of pathogen exposure to develop, if it occurs at all. This repeated pathogen exposure and suboptimal immune response coincide with the expansion of a subset of B cells, often termed atypical memory B cells. This subset is present at low levels in healthy individuals as well but it is observed to expand in an inflammatory context during acute and chronic infection, autoimmune diseases or certain immunodeficiencies. Therefore, it has been proposed that this subset is exhausted, dysfunctional, or potentially autoreactive, but its actual role has remained elusive. Recent reports have provided new information regarding both heterogeneity and expansion of these cells, in addition to indications on their potential role during normal immune responses to infection or vaccination. These new insights encourage us to rethink how and why they are generated and better understand their role in our complex immune system. In this review, we will focus on recent advances in our understanding of these enigmatic cells and highlight the remaining gaps that need to be filled.
Collapse
Affiliation(s)
- Alan-Dine Courey-Ghaouzi
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Linn Kleberg
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- *Correspondence: Christopher Sundling,
| |
Collapse
|
46
|
Seals MR, Moran MM, Leavenworth JD, Leavenworth JW. Contribution of Dysregulated B-Cells and IgE Antibody Responses to Multiple Sclerosis. Front Immunol 2022; 13:900117. [PMID: 35784370 PMCID: PMC9243362 DOI: 10.3389/fimmu.2022.900117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS), a debilitating autoimmune inflammatory disease that affects the brain and spinal cord, causes demyelination of neurons, axonal damage, and neurodegeneration. MS and the murine experimental autoimmune encephalomyelitis (EAE) model have been viewed mainly as T-cell-mediated diseases. Emerging data have suggested the contribution of B-cells and autoantibodies to the disease progression. However, the underlying mechanisms by which dysregulated B-cells and antibody response promote MS and EAE remain largely unclear. Here, we provide an updated review of this specific subject by including B-cell biology and the role of B-cells in triggering autoimmune neuroinflammation with a focus on the regulation of antibody-producing B-cells. We will then discuss the role of a specific type of antibody, IgE, as it relates to the potential regulation of microglia and macrophage activation, autoimmunity and MS/EAE development. This knowledge can be utilized to develop new and effective therapeutic approaches to MS, which fits the scope of the Research Topic "Immune Mechanism in White Matter Lesions: Clinical and Pathophysiological Implications".
Collapse
Affiliation(s)
- Malik R. Seals
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Multidisciplinary Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Monica M. Moran
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jonathan D. Leavenworth
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- The O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
47
|
Rojas F, Parra ER, Wistuba II, Haymaker C, Solis Soto LM. Pathological Response and Immune Biomarker Assessment in Non-Small-Cell Lung Carcinoma Receiving Neoadjuvant Immune Checkpoint Inhibitors. Cancers (Basel) 2022; 14:cancers14112775. [PMID: 35681755 PMCID: PMC9179283 DOI: 10.3390/cancers14112775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Recently, the U.S. Food and Drug Administration (FDA) approved neoadjuvant immunotherapy plus chemotherapy for the treatment of resectable non-small-cell lung carcinoma (NSCLC) due to the clinical benefits reported in several clinical trials. In these settings, the pathological assessment of the tumor bed to quantify a pathological response has been used as a surrogate method of clinical benefit to neoadjuvant therapy. In addition, several clinical trials are including the assessment of tissue-, blood-, or host-based biomarkers to predict therapy response and to monitor the response to neoadjuvant treatment. In this manuscript, we provide an overview of current recommendations for the evaluation of pathological response and describe potential biomarkers used in clinical trials of neoadjuvant immunotherapy in resectable NSCLC. Abstract Lung cancer is the leading cause of cancer incidence and mortality worldwide. Adjuvant and neoadjuvant chemotherapy have been used in the perioperative setting of non-small-cell carcinoma (NSCLC); however, the five-year survival rate only improves by about 5%. Neoadjuvant treatment with immune checkpoint inhibitors (ICIs) has become significant due to improved survival in advanced NSCLC patients treated with immunotherapy agents. The assessment of pathology response has been proposed as a surrogate indicator of the benefits of neaodjuvant therapy. An outline of recommendations has been published by the International Association for the Study of Lung Cancer (IASLC) for the evaluation of pathologic response (PR). However, recent studies indicate that evaluations of immune-related changes are distinct in surgical resected samples from patients treated with immunotherapy. Several clinical trials of neoadjuvant immunotherapy in resectable NSCLC have included the study of biomarkers that can predict the response of therapy and monitor the response to treatment. In this review, we provide relevant information on the current recommendations of the assessment of pathological responses in surgical resected NSCLC tumors treated with neoadjuvant immunotherapy, and we describe current and potential biomarkers to predict the benefits of neoadjuvant immunotherapy in patients with resectable NSCLC.
Collapse
|
48
|
Atkinson BK, Goddard A, Engelbrecht M, Pretorius S, Pazzi P. Circulating markers of endothelial activation in canine parvoviral enteritis. J S Afr Vet Assoc 2022. [DOI: 10.36303/jsava.2022.93.1.496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Affiliation(s)
- BK Atkinson
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria,
South Africa
| | - A Goddard
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria,
South Africa
| | - M Engelbrecht
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria,
South Africa
| | - S Pretorius
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria,
South Africa
| | - P Pazzi
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria,
South Africa
| |
Collapse
|
49
|
Ahmad Zamri N, Rusli MEF, Mohamad Yusof L, Rosli R. Immunization with a bicistronic DNA vaccine modulates systemic IFN-γ and IL-10 expression against Vibrio cholerae infection. J Med Microbiol 2022; 71. [PMID: 35635780 DOI: 10.1099/jmm.0.001536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Cholera is an acute enteric infection caused by Vibrio cholerae, particularly in areas lacking access to clean water. Despite the global effort to improve water quality in these regions, the burden of cholera in recent years has not yet declined. Interest has therefore extended in the use of bicistronic DNA vaccine encoding ctxB and tcpA genes of V. cholerae as a potential vaccine.Hypothesis/Gap Statement. The potential of a bicistronic DNA vaccine, pVAX-ctxB-tcpA has not been determined in vitro and in vivo.Aim. The goal of present study was to evaluate in vitro expression and in vivo potential of pVAX-ctxB-tcpA vaccine against V. cholerae.Methodology. The pVAX-ctxB-tcpA was transiently transfected into mammalian COS-7 cells, and the in vitro expression was assessed using fluorescence and Western blot analyses. Next, the vaccine was encapsulated into sodium alginate using water-in-oil emulsification and evaluated for its efficiency in different pH conditions. Subsequently, oral vaccination using en(pVAX-ctxB-tcpA) was performed in vivo. The animals were challenged with V. cholerae O1 El Tor after 2 weeks of vaccination using the Removable Intestinal Tie-Adult Rabbit Diarrhoea (RITARD) model. Following the infection challenge, the rabbits were monitored for evidence of symptoms, and analysed for systemic cytokine expression level (TNF-α, IFN-γ, IL-6 and IL-10) using quantitative real-time polymerase chain reaction.Results. The in vitro expression of pVAX-ctxB-tcpA was successfully verified via fluorescence and Western blot analyses. Meanwhile, in vivo analysis demonstrated that the en(pVAX-ctxB-tcpA) was able to protect the RITARD model against V. cholerae infection due to a lack of evidence on the clinical manifestations of cholera following bacterial challenge. Furthermore, the bicistronic group showed an upregulation of systemic IFN-γ and IL-10 following 12 days of vaccination, though not significant, suggesting the possible activation of both T-helper 1 and 2 types of response. However, upon bacterial challenge, the gene expression of all cytokines did not change.Conclusion. Our findings suggest that the bicistronic plasmid DNA vaccine, pVAX-ctxB-tcpA, showed a potential role in inducing immune response against cholera through upregulation of in vitro gene and protein expression as well as in vivo cytokine gene expression, particularly IFN-γ and IL-10.
Collapse
Affiliation(s)
- Najwa Ahmad Zamri
- Medical Genetics Laboratory, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Muhammad Ehsan Fitri Rusli
- Medical Genetics Laboratory, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Loqman Mohamad Yusof
- Department of Companion Animal and Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Rozita Rosli
- Medical Genetics Laboratory, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
50
|
Bruss C, Kellner K, Ortmann O, Seitz S, Brockhoff G, Hutchinson JA, Wege AK. Advanced Immune Cell Profiling by Multiparameter Flow Cytometry in Humanized Patient-Derived Tumor Mice. Cancers (Basel) 2022; 14:2214. [PMID: 35565343 PMCID: PMC9103756 DOI: 10.3390/cancers14092214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/07/2022] Open
Abstract
"Humanized" mice have been widely used for the characterization of human cancer progression and as a powerful preclinical model. Standardization of multicolor phenotyping could help to identify immune cell patterns involved in checkpoint-related complications. Therefore, we applied established protocols for immune cell profiling to our humanized Patient-Derived Xenograft (hPDX) model. hPDX are characterized by the co-existence of a human immune system and a patient-derived tumor transplant. These mice possess a human-like immune system after CD34+ stem cell transplantation while the reconstitution level of the immune system was not related to the quantity of transplanted CD34+ cells. Contamination ≤ 1.2% by CD3+ cells in the hematopoietic stem cell (HSC) transplant did not trigger abnormal T cell maturation. Different B and T cell differentiation stages were identified, as well as regulatory T cells (Tregs) and exhausted T cells that expressed TIGIT, PD-1, or KLRG1. Overall, the application of standardized protocols for the characterization of immune cells using flow cytometry will contribute to a better understanding of immune-oncologic processes.
Collapse
Affiliation(s)
- Christina Bruss
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Kerstin Kellner
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - James A. Hutchinson
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Anja Kathrin Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| |
Collapse
|