scholarly journals Costimulation of TLR8 responses by CXCL4 in Human Monocytes Mediated by TBK1-IRF5 Signaling and Epigenomic Remodeling

Author(s):  
Lionel B. Ivashkiv ◽  
Chao Yang ◽  
Mahesh Bachu ◽  
Caroline Brauner ◽  
Ruoxi Yuan ◽  
...  

Abstract CXCL4 regulates responses of immune cells to endosomal TLRs and has been implicated in the pathogenesis of inflammatory and fibrotic diseases. However, mechanisms by which CXCL4 modulates TLR responses, and its functions in monocytes/macrophages, are still unclear. Here we report that CXCL4 changes the profile of the TLR8 response in human monocytes by selectively and dramatically amplifying inflammatory gene transcription and IL-1β production while partially attenuating the IFN response. Mechanistically, costimulation by CXCL4 and TLR8 synergistically activated TBK1/IKKε and repurposed these kinases towards an inflammatory response via coupling with IRF5, and by activating the NLRP3 inflammasome without the need for a second signal. CXCL4 strongly induced chromatin remodeling in a cooperative and synergistic manner with TLR8 signaling, inducing de novo enhancers associated with inflammatory genes. These findings identify signaling and epigenomic mechanisms that underly synergistic activation of inflammatory genes by CXCL4 and TLR8, provide a new paradigm for modulation of TLR responses that is relevant for cytokine storm, and suggest targeting the TBK1/IKKε-IRF5 axis may be beneficial in inflammatory diseases.

Cells ◽  
2021 ◽  
Vol 10 (7) ◽  
pp. 1585
Author(s):  
Annamaria Paolini ◽  
Rebecca Borella ◽  
Sara De Biasi ◽  
Anita Neroni ◽  
Marco Mattioli ◽  
...  

Cell death mechanisms are crucial to maintain an appropriate environment for the functionality of healthy cells. However, during viral infections, dysregulation of these processes can be present and can participate in the pathogenetic mechanisms of the disease. In this review, we describe some features of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and some immunopathogenic mechanisms characterizing the present coronavirus disease (COVID-19). Lymphopenia and monocytopenia are important contributors to COVID-19 immunopathogenesis. The fine mechanisms underlying these phenomena are still unknown, and several hypotheses have been raised, some of which assign a role to cell death as far as the reduction of specific types of immune cells is concerned. Thus, we discuss three major pathways such as apoptosis, necroptosis, and pyroptosis, and suggest that all of them likely occur simultaneously in COVID-19 patients. We describe that SARS-CoV-2 can have both a direct and an indirect role in inducing cell death. Indeed, on the one hand, cell death can be caused by the virus entry into cells, on the other, the excessive concentration of cytokines and chemokines, a process that is known as a COVID-19-related cytokine storm, exerts deleterious effects on circulating immune cells. However, the overall knowledge of these mechanisms is still scarce and further studies are needed to delineate new therapeutic strategies.


2013 ◽  
Vol 210 (7) ◽  
pp. 1433-1445 ◽  
Author(s):  
Nataša Obermajer ◽  
Jeffrey L. Wong ◽  
Robert P. Edwards ◽  
Kong Chen ◽  
Melanie Scott ◽  
...  

Nitric oxide (NO) is a ubiquitous mediator of inflammation and immunity, involved in the pathogenesis and control of infectious diseases, autoimmunity, and cancer. We observed that the expression of nitric oxide synthase-2 (NOS2/iNOS) positively correlates with Th17 responses in patients with ovarian cancer (OvCa). Although high concentrations of exogenous NO indiscriminately suppress the proliferation and differentiation of Th1, Th2, and Th17 cells, the physiological NO concentrations produced by patients’ myeloid-derived suppressor cells (MDSCs) support the development of RORγt(Rorc)+IL-23R+IL-17+ Th17 cells. Moreover, the development of Th17 cells from naive-, memory-, or tumor-infiltrating CD4+ T cells, driven by IL-1β/IL-6/IL-23/NO-producing MDSCs or by recombinant cytokines (IL-1β/IL-6/IL-23), is associated with the induction of endogenous NOS2 and NO production, and critically depends on NOS2 activity and the canonical cyclic guanosine monophosphate (cGMP)–cGMP-dependent protein kinase (cGK) pathway of NO signaling within CD4+ T cells. Inhibition of NOS2 or cGMP–cGK signaling abolishes the de novo induction of Th17 cells and selectively suppresses IL-17 production by established Th17 cells isolated from OvCa patients. Our data indicate that, apart from its previously recognized role as an effector mediator of Th17-associated inflammation, NO is also critically required for the induction and stability of human Th17 responses, providing new targets to manipulate Th17 responses in cancer, autoimmunity, and inflammatory diseases.


2010 ◽  
Vol 30 (19) ◽  
pp. 4644-4655 ◽  
Author(s):  
Zhiqiang Du ◽  
Emily T. Crow ◽  
Hyun Seok Kang ◽  
Liming Li

ABSTRACT We have recently reported that the yeast chromatin-remodeling factor Swi1 can exist as a prion, [SWI +], demonstrating a link between prionogenesis and global transcriptional regulation. To shed light on how the Swi1 conformational switch influences Swi1 function and to define the sequence and structural requirements for [SWI +] formation and propagation, we functionally dissected the Swi1 molecule. We show here that the [SWI +] prion features are solely attributable to the first 327 amino acid residues (N), a region that is asparagine rich. N was aggregated in [SWI+ ] cells but diffuse in [swi− ] cells; chromosomal deletion of the N-coding region resulted in [SWI +] loss, and recombinant N peptide was able to form infectious amyloid fibers in vitro, enabling [SWI +] de novo formation through a simple transformation. Although the glutamine-rich middle region (Q) was not sufficient to aggregate in [SWI +] cells or essential for SWI/SNF function, it significantly modified the Swi1 aggregation pattern and Swi1 function. We also show that excessive Swi1 incurred Li+/Na+ sensitivity and that the N/Q regions are important for this gain of sensitivity. Taken together, our results provide the final proof of “protein-only” transmission of [SWI +] and demonstrate that the widely distributed “dispensable” glutamine/asparagine-rich regions/motifs might have important and divergent biological functions.


Author(s):  
Icely PA ◽  
◽  
Vigezzi C ◽  
Rodriguez E ◽  
Miró MS ◽  
...  

Phagocytes, including monocytes/macrophages, play an important role in the host defense during Candida albicans infections. In the L-arginine metabolism, the balance between the activation of two enzymes, inducible Nitric Oxide Synthase (iNOS) and arginase, promotes in the macrophages two alternative metabolic states, while M1 profile is related with host protection, M2 favored the fungal growth and evasion. Our aim was to evaluate the effect of Amphotericin B (AMB) and Fluconazole (FLC) on polarization of human monocytes to M2 profile induced by C. albicans. The human monocytic (Mo) cell line U937 was co-cultured with viable yeast of C. albicans, or Lipopolysaccharides (LPS) or Phorbol-12-myristate-13-acetate (PMA). Nitric Oxide (NO), cytokines production and arginase activity were evaluated. The effect of AMB or FLC on these metabolic pathways in immune cells and on fungus intrinsic arginase activity was studied. C. albicans inhibits NO production in human-monocyte and induces strong host arginase activity (p<0.0001). AMB and FLC inhibited C. albicansinduced arginase activity in immune cells (p<0.001), reaching a percentage of inhibition of 90% for AMB and 78% for FLC. Arginase intrinsic activity of the fungus was blocked by nor-NOHA (arginase inhibitor) and AMB (p<0.05). These results show that C. albicans drives human Mo toward M2 profile and that both antifungal drugs evaluated have the ability to revert C. albicans-induced M2 profile. In a relevant manner, it also provides data about additional effect of AMB as inhibitor of C. albicans endogenous arginase activity. Here in we provide new evidence for the effect of these drugs over the immune cells and the yeast.


2021 ◽  
pp. ji1901348
Author(s):  
Kathrin Thiem ◽  
Samuel T. Keating ◽  
Mihai G. Netea ◽  
Niels P. Riksen ◽  
Cees J. Tack ◽  
...  

Author(s):  
Sema Ketenci ◽  
Şükrü Aynacıoğlu

The current coronavirus disease 2019 (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has emerged in Wuhan, China and has rapidly become global challenges, creating major challenges to health systems in almost every country in the world it has turned into a pandemic. COVID-19 poses a risky clinical situation that can range from mild illness to severe respiratory failure requiring admission to intensive care. It is known to cause cytokine storm in some critically ill patients. However, more and more evidence showed that there is a dramatic increase in cytokine levels in patients diagnosed with COVID-19. Midkine (MK) is involved in various physiological and pathological processes, which some of them are desired and beneficial such as controlling tissue repair and antimicrobial effects, but some others are harmful such as promoting inflammation, carcinogenesis and chemo-resistance. Also, MK is expressed in inflammatory cells and released by endothelial cells under hypoxic conditions. Considering all this information, there are strong data that MK, an important cytokine known to increase in inflammatory diseases, may overexpressed in patients who are positive for COVID-19. The overexpression of MK reveals a picture leading to fibrosis in the lung damage. Therefore, questions arise about how the concentration of MK changes in CoVID-19 patients and can we use it as an inflammation biomarker or in the treatment protocol in the future.


2021 ◽  
Author(s):  
Victoria Emily Barbosa Hipolito

Cells are exposed to diverse extracellular and intracellular cues, and coopt subcellular responses depending on their cellular state and functional demand; including upregulating signalling pathways or adapting organelle function and physiology. The immune system is a tightly regulated cohort of specialized cells with heterogeneous functions. Phagocytes, a type of immune cell, are challenged with disparate environmental stimuli and can adapt intracellularly to promote immunity. Due to their cellular plasticity, we aim to understand the molecular machinery that controls organelle identity and adaptation in immune cells, when challenged with immunostimulatory agents. First, we used a long tubular phagocytic cup, which provides the spatiotemporal resolution necessary to study the stages of phagocytosis. Using this model, we observed the sequential recruitment of early and late endolysosomal markers to the growing cup. Surprisingly, the early endosomal lipid, phosphatidylinositol-3-phosphate [PtdIns(3)P] persisted. We determined a novel pH-based mechanism that induces the dissociation of the Vps34 Class III phosphatidylinositol-3- kinase from tubular cups as they progressively acidify, when reaching 20 µm in length or upon phagosome closure. The detachment of Vps34 stops the production of PtdIns(3)P, allowing for its turnover by PIKfyve. Given that PtdIns(3)P dependent signalling is important for multiple cellular pathways, this mechanism for pH-dependent regulation of Vps34 could be at the center of many PtdIns(3)P-dependent cellular processes. Additionally, we examined how lysosomes, a kingpin organelle essential for pathogen killing, and antigen processing and presentation, adapt in response to phagocyte activation. During phagocyte activation, lysosomes are remodelled from dozens of globular structures to a tubular network, in a process that requires the PI3K-AKT-mTOR signalling pathway. We showed that lysosome tubulation is coupled with an increase in volume and holding capacity. Lysosome remodelling was dependent on de novo synthesis of lysosomal proteins, but independent of TFEB and TFE3 transcription factors, known to scale-up lysosome biogenesis. We demonstrate a novel mechanism of acute organelle expansion via mTORC1-S6K-4E-BP-dependent increase in lysosomal mRNA translation. This process was necessary for efficient and rapid antigen presentation to T-cells by dendritic cells (DCs). Moreover, lysosome remodelling was conserved in DCs activated with select adjuvants, additives used in vaccines to boost efficacy, providing evidence for its possible clinical applicability. Together, we have identified two novel mechanisms controlling organelle identity and adaptation in immune cells.


2021 ◽  
Author(s):  
Victoria Emily Barbosa Hipolito

Cells are exposed to diverse extracellular and intracellular cues, and coopt subcellular responses depending on their cellular state and functional demand; including upregulating signalling pathways or adapting organelle function and physiology. The immune system is a tightly regulated cohort of specialized cells with heterogeneous functions. Phagocytes, a type of immune cell, are challenged with disparate environmental stimuli and can adapt intracellularly to promote immunity. Due to their cellular plasticity, we aim to understand the molecular machinery that controls organelle identity and adaptation in immune cells, when challenged with immunostimulatory agents. First, we used a long tubular phagocytic cup, which provides the spatiotemporal resolution necessary to study the stages of phagocytosis. Using this model, we observed the sequential recruitment of early and late endolysosomal markers to the growing cup. Surprisingly, the early endosomal lipid, phosphatidylinositol-3-phosphate [PtdIns(3)P] persisted. We determined a novel pH-based mechanism that induces the dissociation of the Vps34 Class III phosphatidylinositol-3- kinase from tubular cups as they progressively acidify, when reaching 20 µm in length or upon phagosome closure. The detachment of Vps34 stops the production of PtdIns(3)P, allowing for its turnover by PIKfyve. Given that PtdIns(3)P dependent signalling is important for multiple cellular pathways, this mechanism for pH-dependent regulation of Vps34 could be at the center of many PtdIns(3)P-dependent cellular processes. Additionally, we examined how lysosomes, a kingpin organelle essential for pathogen killing, and antigen processing and presentation, adapt in response to phagocyte activation. During phagocyte activation, lysosomes are remodelled from dozens of globular structures to a tubular network, in a process that requires the PI3K-AKT-mTOR signalling pathway. We showed that lysosome tubulation is coupled with an increase in volume and holding capacity. Lysosome remodelling was dependent on de novo synthesis of lysosomal proteins, but independent of TFEB and TFE3 transcription factors, known to scale-up lysosome biogenesis. We demonstrate a novel mechanism of acute organelle expansion via mTORC1-S6K-4E-BP-dependent increase in lysosomal mRNA translation. This process was necessary for efficient and rapid antigen presentation to T-cells by dendritic cells (DCs). Moreover, lysosome remodelling was conserved in DCs activated with select adjuvants, additives used in vaccines to boost efficacy, providing evidence for its possible clinical applicability. Together, we have identified two novel mechanisms controlling organelle identity and adaptation in immune cells.


2021 ◽  
Vol 4 (1) ◽  
pp. 17-20
Author(s):  
Nazanin Ashtar Nakhaei ◽  
◽  
Amir Hossein Norooznezhad ◽  

Coronavirus Disease 2019 (COVID-19) is one of the most critical health issues in the world. According to the findings, systemic proinflammatory cytokine release is associated with the pathogenesis of cytokine storm, contributing to morbidities and even mortality in patients diagnosed with COVID-19. Among pregnant patients diagnosed with COVID-19, preterm labor is one of the most crucial side effects, with a prevalence of up to 63.8% in some studies. As well as cytokine storm, proinflammatory cytokines are involved in preterm labor. Mesenchymal Stem Cells (MSCs) transplantation has been used in different trials to suppress inflammation in many inflammatory diseases. MSCs have also been successfully applied to treat patients diagnosed with COVID-19, considering the cytokine storm in these patients. So, it is possible to use the transplantation of MSCs derived from the maternal side of the placenta as an autologous product to suppress cytokine storm in critically ill patients diagnosed with COVID-19. The autologous transplantation of MSCs helps to suppress cytokine storm and systemic inflammation. Inhibition of systemic cytokine release could prevent poor outcomes, especially mortality and morbidities in the mentioned patients.


2001 ◽  
Vol 95 (2) ◽  
pp. 500-508 ◽  
Author(s):  
Heike L. Rittner ◽  
Alexander Brack ◽  
Halina Machelska ◽  
Shaaban A. Mousa ◽  
Monika Bauer ◽  
...  

Background Inflammatory pain can be effectively controlled by an interaction of opioid receptors on peripheral sensory nerve terminals with opioid peptides released from immune cells upon stressful stimulation. To define the source of opioid peptide production, we sought to identify and quantify populations of opioid-containing cells during the course of Freund's complete adjuvant-induced hind paw inflammation in the rat. In parallel, we examined the development of stress-induced local analgesia in the paw. Methods At 2, 6, and 96 h after Freund's complete adjuvant inoculation, cells were characterized by flow cytometry using a monoclonal pan-opioid antibody (3E7) and antibodies against cell surface antigens and by immunohistochemistry using a polyclonal antibody to beta-endorphin. After magnetic cell sorting, the beta-endorphin content was quantified by radioimmunoassay. Pain responses before and after cold water swim stress were evaluated by paw pressure thresholds. Results In early inflammation, 66% of opioid peptide-producing (3E7+) leukocytes were HIS48+ granulocytes. In contrast, at later stages (96 h), the majority of 3E7+ immune cells were ED1+ monocytes or macrophages (73%). During the 4 days after Freund's complete adjuvant inoculation, the number of 3E7+ cells increased 5.6-fold (P &lt; 0.001, Kruskal-Wallis test) and the beta-endorphin content in the paw multiplied 3.9-fold (P &lt; 0.05, Kruskal-Wallis test). In parallel, cold water swim stress-induced analgesia increased by 160% (P &lt; 0.01, analysis of variance). Conclusions The degree of endogenous pain inhibition is proportional to the number of opioid peptide-producing cells, and distinct leukocyte lineages contribute to this function at different stages of inflammation. These mechanisms may be important for understanding pain in immunosuppressed states such as cancer, diabetes, or AIDS and for the design of novel therapeutic strategies in inflammatory diseases.


Sign in / Sign up

Export Citation Format

Share Document