ijms-logo

Journal Browser

Journal Browser

Advances in the Pathogenesis and Treatment of Immune-Mediated Inflammatory Diseases

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Immunology".

Deadline for manuscript submissions: closed (30 September 2021) | Viewed by 56422

Special Issue Editors


E-Mail Website
Guest Editor
Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
Interests: immune-mediated inflammatory diseases; molecular regulation of inflammation; signal transduction; B cells; dendritic cells; endothelial cells/angiogenesis

E-Mail Website
Guest Editor
Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
Interests: immune-mediated inflammatory diseases; molecular regulation of inflammation; lymphocytes; stromal cells; NF-κB signaling

Special Issue Information

Dear Colleagues,

This Special Issue will focus on the rapidly evolving field of immune-mediated inflammatory diseases (IMIDs) and the achievements that were made over the last 10 years. With the advent of novel technologies such as single-cell RNA sequencing and state-of-the-art imaging techniques, researchers in our field have made tremendous progress in unraveling the pathogenesis of autoimmune diseases such as rheumatoid arthritis, vasculitis, and systemic lupus erythematosus, as well as autoinflammatory diseases such as spondyloarthritis and fever syndromes. This has also provided new insights into the contribution of specific (subsets of) immune cells and stromal cells to the inflammatory response, as well as the molecular pathways associated with the pathological processes involved. These advances have led to the identification of promising new therapeutic targets for these diseases and paved the way for novel treatments. However, for rare IMIDs, we are far from fully understanding the underlying mechanisms leading to the development and progression of disease, as well as the response to therapy. A better understanding of the molecular and cellular signatures of these diseases may inform us about potential new treatment options that could be explored.

This Special Issue of IJMS will focus on the molecular, cellular, and clinical advances in the pathogenesis and treatment of IMIDs. We will consider original research, review, or methods manuscripts related to this subject and the keywords below.

Dr. Sander W. Tas
Dr. Jan Piet van Hamburg
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • autoimmunity
  • inflammation
  • immune cells
  • stromal cells
  • lymphoid organs
  • inflammatory mediators
  • signal transduction
  • state-of-the-art technology, including (sc)RNAseq and (imaging)CyTOF
  • molecular imaging and advanced microscopy
  • animal models

Published Papers (15 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

3 pages, 176 KiB  
Editorial
Editorial for Special Issue “Advances in the Pathogenesis and Treatment of Immune-Mediated Inflammatory Diseases”
by Jan Piet van Hamburg and Sander W. Tas
Int. J. Mol. Sci. 2022, 23(15), 8415; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158415 - 29 Jul 2022
Viewed by 798
Abstract
This Special Issue focuses on the rapidly evolving field of immune-mediated inflammatory diseases (IMIDs) and the achievements that were made over the last 10 years [...] Full article

Research

Jump to: Editorial, Review

13 pages, 1454 KiB  
Article
Decreased BAFF Receptor Expression and Unaltered B Cell Receptor Signaling in Circulating B Cells from Primary Sjögren’s Syndrome Patients at Diagnosis
by Stefan F. H. Neys, Gwenny M. Verstappen, Hendrika Bootsma, Frans G. M. Kroese, Rudi W. Hendriks and Odilia B. J. Corneth
Int. J. Mol. Sci. 2022, 23(9), 5101; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23095101 - 04 May 2022
Cited by 4 | Viewed by 2254
Abstract
Animal models of autoimmunity and human genetic association studies indicate that the dysregulation of B-cell receptor (BCR) signaling is an important driver of autoimmunity. We previously showed that in circulating B cells from primary Sjögren’s syndrome (pSS) patients with high systemic disease activity, [...] Read more.
Animal models of autoimmunity and human genetic association studies indicate that the dysregulation of B-cell receptor (BCR) signaling is an important driver of autoimmunity. We previously showed that in circulating B cells from primary Sjögren’s syndrome (pSS) patients with high systemic disease activity, protein expression of the BCR signaling molecule Bruton’s tyrosine kinase (BTK) was increased and correlated with T-cell infiltration in the target organ. We hypothesized that these alterations could be driven by increased B-cell activating factor (BAFF) levels in pSS. Here, we investigated whether altered BCR signaling was already present at diagnosis and distinguished pSS from non-SS sicca patients. Using (phospho-)flow cytometry, we quantified the phosphorylation of BCR signaling molecules, and investigated BTK and BAFF receptor (BAFFR) expression in circulating B cell subsets in an inception cohort of non-SS sicca and pSS patients, as well as healthy controls (HCs). We found that both BTK protein levels and BCR signaling activity were comparable among groups. Interestingly, BAFFR expression was significantly downregulated in pSS, but not in non-SS sicca patients, compared with HCs, and correlated with pSS-associated alterations in B cell subsets. These data indicate reduced BAFFR expression as a possible sign of early B cell involvement and a diagnostic marker for pSS. Full article
Show Figures

Figure 1

16 pages, 3357 KiB  
Article
Splenic Architecture and Function Requires Tight Control of Transmembrane TNF Expression
by Kim C. M. Jeucken, Merlijn H. Kaaij, Jasper Rip, Charlotte C. N. van Rooijen, Yik Y. Kan, Odilia B. J. Corneth, Jan Piet van Hamburg and Sander W. Tas
Int. J. Mol. Sci. 2022, 23(4), 2229; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23042229 - 17 Feb 2022
Cited by 3 | Viewed by 2037
Abstract
Soluble tumor necrosis factor (sTNF) is an important inflammatory mediator and essential for secondary lymphoid organ (SLO) development and function. However, the role of its transmembrane counterpart (tmTNF) in these processes is less well established. Here, the effects of tmTNF overxpression on SLO [...] Read more.
Soluble tumor necrosis factor (sTNF) is an important inflammatory mediator and essential for secondary lymphoid organ (SLO) development and function. However, the role of its transmembrane counterpart (tmTNF) in these processes is less well established. Here, the effects of tmTNF overxpression on SLO architecture and function were investigated using tmTNF-transgenic (tmTNF-tg) mice. tmTNF overexpression resulted in enlarged peripheral lymph nodes (PLNs) and spleen, accompanied by an increase in small splenic lymphoid follicles, with less well-defined primary B cell follicles and T cell zones. In tmTNF-tg mice, the spleen, but not PLNs, contained reduced germinal center (GC) B cell fractions, with low Ki67 expression and reduced dark zone characteristics. In line with this, smaller fractions of T follicular helper (Tfh) and T follicular regulatory (Tfr) cells were observed with a decreased Tfh:Tfr ratio. Moreover, plasma cell (PC) formation in the spleen of tmTNF-tg mice decreased and skewed towards IgA and IgM expression. Genetic deletion of TNFRI or –II resulted in a normalization of follicle morphology in the spleen of tmTNF-tg mice, but GC B cell and PC fractions remained abnormal. These findings demonstrate that tightly regulated tmTNF is important for proper SLO development and function, and that aberrations induced by tmTNF overexpression are site-specific and mediated via TNFRI and/or TNFRII signaling. Full article
Show Figures

Figure 1

15 pages, 53423 KiB  
Article
Dual Blockade of TNF and IL-17A Inhibits Inflammation and Structural Damage in a Rat Model of Spondyloarthritis
by Ihsan Hammoura, Renee H. Fiechter, Shaughn H. Bryant, Susan Westmoreland, Gillian Kingsbury, Wendy Waegell, Sander W. Tas, Dominique L. Baeten, Marleen G. H. van de Sande, Melissa N. van Tok and Leonie M. van Duivenvoorde
Int. J. Mol. Sci. 2022, 23(2), 859; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23020859 - 13 Jan 2022
Cited by 14 | Viewed by 2795
Abstract
The tumor necrosis factor (TNF) and IL-23/IL-17 axes are the main therapeutic targets in spondyloarthritis. Despite the clinical efficacy of blocking either pathway, monotherapy does not induce remission in all patients and its effect on new bone formation remains unclear. We aimed to [...] Read more.
The tumor necrosis factor (TNF) and IL-23/IL-17 axes are the main therapeutic targets in spondyloarthritis. Despite the clinical efficacy of blocking either pathway, monotherapy does not induce remission in all patients and its effect on new bone formation remains unclear. We aimed to study the effect of TNF and IL-17A dual inhibition on clinical disease and structural damage using the HLA-B27/human β2-microglobulin transgenic rat model of SpA. Immunized rats were randomized according to arthritis severity, 1 week after arthritis incidence reached 50%, to be treated twice weekly for a period of 5 weeks with either a dual blockade therapy of an anti-TNF antibody and an anti-IL-17A antibody, a single therapy of either antibody, or PBS as vehicle control. Treatment-blinded observers assessed inflammation and structural damage clinically, histologically and by micro-CT imaging. Both single therapies as well as TNF and IL-17A dual blockade therapy reduced clinical spondylitis and peripheral arthritis effectively and similarly. Clinical improvement was confirmed for all treatments by a reduction of histological inflammation and pannus formation (p < 0.05) at the caudal spine. All treatments showed an improvement of structural changes at the axial and peripheral joints on micro-CT imaging, with a significant decrease for roughness (p < 0.05), which reflects both erosion and new bone formation, at the level of the caudal spine. The effect of dual blockade therapy on new bone formation was more prominent at the axial than the peripheral level. Collectively, our study showed that dual blockade therapy significantly reduces inflammation and structural changes, including new bone formation. However, we could not confirm a more pronounced effect of dual inhibition compared to single inhibition. Full article
Show Figures

Figure 1

13 pages, 2545 KiB  
Article
Fibroblast Activation Protein Targeted Photodynamic Therapy Selectively Kills Activated Skin Fibroblasts from Systemic Sclerosis Patients and Prevents Tissue Contraction
by Daphne N. Dorst, Arjan P. M. van Caam, Elly L. Vitters, Birgitte Walgreen, Monique M. A. Helsen, Christian Klein, Shreya Gudi, Tirza Wubs, Jyoti Kumari, Madelon C. Vonk, Peter M. van der Kraan and Marije I. Koenders
Int. J. Mol. Sci. 2021, 22(23), 12681; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222312681 - 24 Nov 2021
Cited by 9 | Viewed by 2787
Abstract
Systemic sclerosis (SSc) is a rare, severe, auto-immune disease characterized by inflammation, vasculopathy and fibrosis. Activated (myo)fibroblasts are crucial drivers of this fibrosis. By exploiting their expression of fibroblast activation protein (FAP) to perform targeted photodynamic therapy (tPDT), we can locoregionally deplete these [...] Read more.
Systemic sclerosis (SSc) is a rare, severe, auto-immune disease characterized by inflammation, vasculopathy and fibrosis. Activated (myo)fibroblasts are crucial drivers of this fibrosis. By exploiting their expression of fibroblast activation protein (FAP) to perform targeted photodynamic therapy (tPDT), we can locoregionally deplete these pathogenic cells. In this study, we explored the use of FAP-tPDT in primary skin fibroblasts from SSc patients, both in 2D and 3D cultures. Method: The FAP targeting antibody 28H1 was conjugated with the photosensitizer IRDye700DX. Primary skin fibroblasts were obtained from lesional skin biopsies of SSc patients via spontaneous outgrowth and subsequently cultured on plastic or collagen type I. For 2D FAP-tPDT, cells were incubated in buffer with or without the antibody-photosensitizer construct, washed after 4 h and exposed to λ = 689 nm light. Cell viability was measured using CellTiter Glo®®. For 3D FAP-tPDT, cells were seeded in collagen plugs and underwent the same treatment procedure. Contraction of the plugs was followed over time to determine myofibroblast activity. Results: FAP-tPDT resulted in antibody-dose dependent cytotoxicity in primary skin fibroblasts upon light exposure. Cells not exposed to light or incubated with an irrelevant antibody-photosensitizer construct did not show this response. FAP-tPDT fully prevented contraction of collagen plugs seeded with primary SSc fibroblasts. Even incubation with a very low dose of antibody (0.4 nM) inhibited contraction in 2 out of 3 donors. Conclusions: Here we have shown, for the first time, the potential of FAP-tPDT for the treatment of fibrosis in SSc skin. Full article
Show Figures

Figure 1

13 pages, 3331 KiB  
Article
Takinib Inhibits Inflammation in Human Rheumatoid Arthritis Synovial Fibroblasts by Targeting the Janus Kinase-Signal Transducer and Activator of Transcription 3 (JAK/STAT3) Pathway
by Paul M. Panipinto, Anil K. Singh, Farheen S. Shaikh, Ruby J. Siegel, Mukesh Chourasia and Salahuddin Ahmed
Int. J. Mol. Sci. 2021, 22(22), 12580; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222212580 - 22 Nov 2021
Cited by 8 | Viewed by 2607
Abstract
TGF β-activated kinase 1 (TAK1) is an important participant in inflammatory pathogenesis for diseases such as rheumatoid arthritis (RA) and gouty arthritis. The central position it occupies between the mitogen activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways makes it [...] Read more.
TGF β-activated kinase 1 (TAK1) is an important participant in inflammatory pathogenesis for diseases such as rheumatoid arthritis (RA) and gouty arthritis. The central position it occupies between the mitogen activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways makes it an attractive therapeutic target. As this field has developed in recent years, several novel inhibitors have been presented as having specific activity that reduces the TAK1 function either covalently as in the case of 5Z-7-oxozeanol (5Z7O) or reversibly (NG-25). However, the mechanism through which takinib elicits its anti-inflammatory activity remains elusive. While this inhibitor shows great promise, a thorough analysis of its inhibitor function and its potential off-target effects is necessary before addressing its clinical potential or its use in inflammatory conditions. An analysis through Western blot showed an unexpected increase in IL-1β-induced TAK1 phosphorylation—a prerequisite for and indicator of its functional potential—by takinib while simultaneously demonstrating the inhibition of the JAK/STAT pathway in human rheumatoid arthritis synovial fibroblasts (RASFs) in vitro. In THP-1 monocyte-derived macrophages, takinib again led to the lipopolysaccharide-induced phosphorylation of TAK1 without a marked inhibition of the TAK1 downstream effectors, namely, of c-Jun N-terminal kinase (JNK), phospho-c-Jun, NF-κB phospho-p65 or phospho-IκBα. Taken together, these findings indicate that takinib inhibits inflammation in these cells by targeting multiple signaling pathways, most notably the JAK/STAT pathway in human RASFs. Full article
Show Figures

Figure 1

12 pages, 1515 KiB  
Article
C-Reactive Protein Controls IL-23 Production by Human Monocytes
by Chiara E. Geyer, Melissa Newling, Lathees Sritharan, Guillermo R. Griffith, Hung-Jen Chen, Dominique L. P. Baeten and Jeroen den Dunnen
Int. J. Mol. Sci. 2021, 22(21), 11638; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222111638 - 28 Oct 2021
Cited by 6 | Viewed by 2359
Abstract
C-reactive protein (CRP) is an acute-phase protein in humans that is produced in high quantities by the liver upon infection and under inflammatory conditions. Although CRP is commonly used as a marker of inflammation, CRP can also directly contribute to inflammation by eliciting [...] Read more.
C-reactive protein (CRP) is an acute-phase protein in humans that is produced in high quantities by the liver upon infection and under inflammatory conditions. Although CRP is commonly used as a marker of inflammation, CRP can also directly contribute to inflammation by eliciting pro-inflammatory cytokine production by immune cells. Since CRP is highly elevated in serum under inflammatory conditions, we have studied the CRP-induced cytokine profile of human monocytes, one of the main innate immune cell populations in blood. We identified that CRP is relatively unique in its capacity to induce production of the pro-inflammatory cytokine IL-23, which was in stark contrast to a wide panel of pattern recognition receptor (PRR) ligands. We show that CRP-induced IL-23 production was mediated at the level of gene transcription, since CRP particularly promoted gene transcription of IL23A (encoding IL-23p19) instead of IL12A (encoding IL-12p35), while PRR ligands induce the opposite response. Interestingly, when CRP stimulation was combined with PRR ligand stimulation, as for example, occurs in the context of sepsis, IL-23 production by monocytes was strongly reduced. Combined, these data identify CRP as a unique individual ligand to induce IL-23 production by monocytes, which may contribute to shaping systemic immune responses under inflammatory conditions. Full article
Show Figures

Figure 1

19 pages, 3672 KiB  
Article
Efficient Neutrophil Activation Requires Two Simultaneous Activating Stimuli
by Sanne Mol, Florianne M. J. Hafkamp, Laura Varela, Neena Simkhada, Esther W. Taanman-Kueter, Sander W. Tas, Marca H. M. Wauben, Tom Groot Kormelink and Esther C. de Jong
Int. J. Mol. Sci. 2021, 22(18), 10106; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221810106 - 18 Sep 2021
Cited by 25 | Viewed by 4483
Abstract
Neutrophils are abundantly present in the synovium and synovial fluid of patients suffering from arthritis. Neutrophils can be activated by a multitude of stimuli and the current dogma states that this is a two-step process, consisting of a priming step followed by an [...] Read more.
Neutrophils are abundantly present in the synovium and synovial fluid of patients suffering from arthritis. Neutrophils can be activated by a multitude of stimuli and the current dogma states that this is a two-step process, consisting of a priming step followed by an activation step. Considering that neutrophil activation occurs in an inflammatory environment, where multiple stimuli are present, we argue that a two-step process is highly unlikely. Here, we indeed demonstrate that neutrophils require simultaneous ligation of two different receptors for efficient activation. We isolated human peripheral blood neutrophils and cultured them with various combinations of stimuli (GM-CSF, fMLF, TNF, and LPS). Next, we evaluated essential neutrophil functions, including degranulation and ROS production using flow cytometry, mediator release using ELISA, NETosis by a live cell imaging method, phagocytosis by imaging flow cytometry, and extracellular vesicle (EV) release quantified by high-resolution flow cytometry. Exposure of neutrophils to any combination of stimuli, but not to single stimuli, resulted in significant degranulation, and mediator and EV release. Furthermore, ROS production increased substantially by dual stimulation, yet appeared to be more dependent on the type of stimulation than on dual stimulation. Phagocytosis was induced to its maximum capacity by a single stimulus, while NETosis was not induced by any of the used physiological stimuli. Our data indicate that neutrophil activation is tightly regulated and requires activation by two simultaneous stimuli, which is largely independent of the combination of stimuli. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

25 pages, 863 KiB  
Review
Pathophysiological Mechanisms and Treatment of Dermatomyositis and Immune Mediated Necrotizing Myopathies: A Focused Review
by Renske G. Kamperman, Anneke J. van der Kooi, Marianne de Visser, Eleonora Aronica and Joost Raaphorst
Int. J. Mol. Sci. 2022, 23(8), 4301; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23084301 - 13 Apr 2022
Cited by 18 | Viewed by 5671
Abstract
Idiopathic inflammatory myopathies (IIM), collectively known as myositis, are a composite group of rare autoimmune diseases affecting mostly skeletal muscle, although other organs or tissues may also be involved. The main clinical feature of myositis is subacute, progressive, symmetrical muscle weakness in the [...] Read more.
Idiopathic inflammatory myopathies (IIM), collectively known as myositis, are a composite group of rare autoimmune diseases affecting mostly skeletal muscle, although other organs or tissues may also be involved. The main clinical feature of myositis is subacute, progressive, symmetrical muscle weakness in the proximal arms and legs, whereas subtypes of myositis may also present with extramuscular features, such as skin involvement, arthritis or interstitial lung disease (ILD). Established subgroups of IIM include dermatomyositis (DM), immune-mediated necrotizing myopathy (IMNM), anti-synthetase syndrome (ASyS), overlap myositis (OM) and inclusion body myositis (IBM). Although these subgroups have overlapping clinical features, the widespread variation in the clinical manifestations of IIM suggests different pathophysiological mechanisms. Various components of the immune system are known to be important immunopathogenic pathways in IIM, although the exact pathophysiological mechanisms causing the muscle damage remain unknown. Current treatment, which consists of glucocorticoids and other immunosuppressive or immunomodulating agents, often fails to achieve a sustained beneficial response and is associated with various adverse effects. New therapeutic targets have been identified that may improve outcomes in patients with IIM. A better understanding of the overlapping and diverging pathophysiological mechanisms of the major subgroups of myositis is needed to optimize treatment. The aim of this review is to report on recent advancements regarding DM and IMNM. Full article
Show Figures

Figure 1

20 pages, 1431 KiB  
Review
The Role of the Lymphatic System in the Pathogenesis and Treatment of Inflammatory Bowel Disease
by Dimitrios Nikolakis, Floris A. E. de Voogd, Maarten J. Pruijt, Joep Grootjans, Marleen G. van de Sande and Geert R. D’Haens
Int. J. Mol. Sci. 2022, 23(3), 1854; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23031854 - 06 Feb 2022
Cited by 14 | Viewed by 4922
Abstract
Although the number of therapeutic options for the treatment of inflammatory bowel disease (IBD) has increased in recent years, patients suffer from decreased quality of life due to non-response or loss of response to the currently available treatments. An increased understanding of the [...] Read more.
Although the number of therapeutic options for the treatment of inflammatory bowel disease (IBD) has increased in recent years, patients suffer from decreased quality of life due to non-response or loss of response to the currently available treatments. An increased understanding of the disease’s etiology could provide novel insights for treatment strategies in IBD. Lymphatic system components are generally linked to immune responses and presumably related to inflammatory diseases pathophysiology. This review aims to summarize findings on immune-mediated mechanisms in lymphoid tissues linked with IBD pathogenesis and (potential) novel treatments. Enhanced innate and adaptive immune responses were observed in mesenteric lymph nodes (MLNs) and other lymphoid structures, such as Peyer’s patches, in patients with IBD and in animal models. Furthermore, the phenomenon of lymphatic obstruction in the form of granulomas in MLNs and lymphatic vessels correlates with disease activity. There is also evidence that abnormalities in the lymphatic stromal components and lymph node microbiome are common in IBD and could be exploited therapeutically. Finally, novel agents targeting lymphocyte trafficking have been added to the treatment armamentarium in the field of IBD. Overall, gut-associated lymphoid tissue plays a key role in IBD immunopathogenesis, which could offer novel therapeutic targets. Full article
Show Figures

Graphical abstract

18 pages, 1271 KiB  
Review
B Lineage Cells in ANCA-Associated Vasculitis
by Ana Merino-Vico, Jan Piet van Hamburg and Sander W. Tas
Int. J. Mol. Sci. 2022, 23(1), 387; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23010387 - 30 Dec 2021
Cited by 12 | Viewed by 3119
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a systemic autoimmune disease that affects small sized blood vessels and can lead to serious complications in the lungs and kidneys. The prominent presence of ANCA autoantibodies in this disease implicates B cells in its pathogenesis, [...] Read more.
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a systemic autoimmune disease that affects small sized blood vessels and can lead to serious complications in the lungs and kidneys. The prominent presence of ANCA autoantibodies in this disease implicates B cells in its pathogenesis, as these are the precursors of the ANCA-producing plasma cells (PCs). Further evidence supporting the potential role of B lineage cells in vasculitis are the increased B cell cytokine levels and the dysregulated B cell populations in patients. Confirmation of the contribution of B cells to pathology arose from the beneficial effect of anti-CD20 therapy (i.e., rituximab) in AAV patients. These anti-CD20 antibodies deplete circulating B cells, which results in amelioration of disease. However, not all patients respond completely, and this treatment does not target PCs, which can maintain ANCA production. Hence, it is important to develop more specific therapies for AAV patients. Intracellular signalling pathways may be potential therapeutic targets as they can show (disease-specific) alterations in certain B lineage cells, including pathogenic B cells, and contribute to differentiation and survival of PCs. Preliminary data on the inhibition of certain signalling molecules downstream of receptors specific for B lineage cells show promising therapeutic effects. In this narrative review, B cell specific receptors and their downstream signalling molecules that may contribute to pathology in AAV are discussed, including the potential to therapeutically target these pathways. Full article
Show Figures

Figure 1

22 pages, 1263 KiB  
Review
B Cell Involvement in the Pathogenesis of Ankylosing Spondylitis
by Rick Wilbrink, Anneke Spoorenberg, Gwenny M. P. J. Verstappen and Frans G. M. Kroese
Int. J. Mol. Sci. 2021, 22(24), 13325; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222413325 - 11 Dec 2021
Cited by 18 | Viewed by 5366
Abstract
Extensive research into ankylosing spondylitis (AS) has suggested the major role of genetics, immune reactions, and the joint–gut axis in its etiology, although an ultimate consensus does not yet exist. The available evidence indicates that both autoinflammation and T-cell-mediated autoimmune processes are actively [...] Read more.
Extensive research into ankylosing spondylitis (AS) has suggested the major role of genetics, immune reactions, and the joint–gut axis in its etiology, although an ultimate consensus does not yet exist. The available evidence indicates that both autoinflammation and T-cell-mediated autoimmune processes are actively involved in the disease process of AS. So far, B cells have received relatively little attention in AS pathogenesis; this is largely due to a lack of conventional disease-defining autoantibodies. However, against prevailing dogma, there is a growing body of evidence suggestive of B cell involvement. This is illustrated by disturbances in circulating B cell populations and the formation of auto-reactive and non-autoreactive antibodies, along with B cell infiltrates within the axial skeleton of AS patients. Furthermore, the depletion of B cells, using rituximab, displayed beneficial results in a subgroup of patients with AS. This review provides an overview of our current knowledge of B cells in AS, and discusses their potential role in its pathogenesis. An overarching picture portrays increased B cell activation in AS, although it is unclear whether B cells directly affect pathogenesis, or are merely bystanders in the disease process. Full article
Show Figures

Figure 1

23 pages, 1675 KiB  
Review
The Role of the Immune System in IBD-Associated Colorectal Cancer: From Pro to Anti-Tumorigenic Mechanisms
by Sofía Frigerio, Dalia A. Lartey, Geert R. D’Haens and Joep Grootjans
Int. J. Mol. Sci. 2021, 22(23), 12739; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222312739 - 25 Nov 2021
Cited by 26 | Viewed by 4891
Abstract
Patients with inflammatory bowel disease (IBD) have increased incidence of colorectal cancer (CRC). IBD-associated cancer follows a well-characterized sequence of intestinal epithelial changes, in which genetic mutations and molecular aberrations play a key role. IBD-associated cancer develops against a background of chronic inflammation [...] Read more.
Patients with inflammatory bowel disease (IBD) have increased incidence of colorectal cancer (CRC). IBD-associated cancer follows a well-characterized sequence of intestinal epithelial changes, in which genetic mutations and molecular aberrations play a key role. IBD-associated cancer develops against a background of chronic inflammation and pro-inflammatory immune cells, and their products contribute to cancer development and progression. In recent years, the effect of the immunosuppressive microenvironment in cancer development and progression has gained more attention, mainly because of the unprecedented anti-tumor effects of immune checkpoint inhibitors in selected groups of patients. Even though IBD-associated cancer develops in the background of chronic inflammation which is associated with activation of endogenous anti-inflammatory or suppressive mechanisms, the potential role of an immunosuppressive microenvironment in these cancers is largely unknown. In this review, we outline the role of the immune system in promoting cancer development in chronic inflammatory diseases such as IBD, with a specific focus on the anti-inflammatory mechanisms and suppressive immune cells that may play a role in IBD-associated tumorigenesis. Full article
Show Figures

Figure 1

16 pages, 1232 KiB  
Review
Single-Cell RNA Sequencing Reveals Heterogeneity and Functional Diversity of Lymphatic Endothelial Cells
by Hannah den Braanker, Astrid C. van Stigt, Marc R. Kok, Erik Lubberts and Radjesh J. Bisoendial
Int. J. Mol. Sci. 2021, 22(21), 11976; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222111976 - 05 Nov 2021
Cited by 9 | Viewed by 5778
Abstract
Lymphatic endothelial cells (LECs) line the lymphatic vasculature and play a central role in the immune response. LECs have abilities to regulate immune transport, to promote immune cell survival, and to cross present antigens to dendritic cells. Single-cell RNA sequencing (scRNA) technology has [...] Read more.
Lymphatic endothelial cells (LECs) line the lymphatic vasculature and play a central role in the immune response. LECs have abilities to regulate immune transport, to promote immune cell survival, and to cross present antigens to dendritic cells. Single-cell RNA sequencing (scRNA) technology has accelerated new discoveries in the field of lymphatic vascular biology. This review will summarize these new findings in regard to embryonic development, LEC heterogeneity with associated functional diversity, and interactions with other cells. Depending on the organ, location in the lymphatic vascular tree, and micro-environmental conditions, LECs feature unique properties and tasks. Furthermore, adjacent stromal cells need the support of LECs for fulfilling their tasks in the immune response, such as immune cell transport and antigen presentation. Although aberrant lymphatic vasculature has been observed in a number of chronic inflammatory diseases, the knowledge on LEC heterogeneity and functional diversity in these diseases is limited. Combining scRNA sequencing data with imaging and more in-depth functional experiments will advance our knowledge of LECs in health and disease. Building the case, the LEC could be put forward as a new therapeutic target in chronic inflammatory diseases, counterweighting the current immune-cell focused therapies. Full article
Show Figures

Figure 1

15 pages, 8147 KiB  
Review
The Germinal Center Milieu in Rheumatoid Arthritis: The Immunological Drummer or Dancer?
by Dornatien C. Anang, Giulia Balzaretti, Antoine van Kampen, Niek de Vries and Paul L. Klarenbeek
Int. J. Mol. Sci. 2021, 22(19), 10514; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910514 - 29 Sep 2021
Cited by 3 | Viewed by 5094
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by joint inflammation, affecting approximately 1% of the general population. To alleviate symptoms and ameliorate joint damage, chronic use of immunosuppressives is needed. However, these treatments are only partially effective and may lead to [...] Read more.
Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by joint inflammation, affecting approximately 1% of the general population. To alleviate symptoms and ameliorate joint damage, chronic use of immunosuppressives is needed. However, these treatments are only partially effective and may lead to unwanted side effects. Therefore, a more profound understanding of the pathophysiology might lead to more effective therapies, or better still, a cure. The presence of autoantibodies in RA indicates that B cells might have a pivotal role in the disease. This concept is further supported by the fact that a diverse antibody response to various arthritis-related epitopes is associated with arthritis development. In this context, attention has focused in recent years on the role of Germinal Centers (GCs) in RA. Since GCs act as the main anatomic location of somatic hypermutations, and, thus, contributing to the diversity and specificity of (auto) antibodies, it has been speculated that defects in germinal center reactions might be crucial in the initiation and maintenance of auto-immune events. In this paper, we discuss current evidence that various processes within GCs can result in the aberrant production of B cells that possess autoreactive properties and might result in the production of RA related autoantibodies. Secondly, we discuss various (pre-)clinical studies that have targeted various GC processes as novel therapies for RA treatment. Full article
Show Figures

Figure 1

Back to TopTop