1
|
Fertitta V, Varano B, Del Cornò M, Fortini P, Aureli A, Conti L. Akkermansia muciniphila- and Pathogenic Bacteria-Derived Endotoxins Differently Regulate Human Dendritic Cell Generation and γδ T Lymphocyte Activation. Biomolecules 2024; 14:1571. [PMID: 39766278 PMCID: PMC11673428 DOI: 10.3390/biom14121571] [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: 10/25/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Lipopolysaccharide (LPS) is a potent endotoxin released at high concentrations in acute infections, causing massive host inflammatory response. Accumulating evidence indicates that dysbiosis-associated chronic low levels of circulating LPS can sustain a prolonged sterile low-grade inflammation that increases the risk of several non-communicable diseases. Interventions aimed at increasing the abundance of beneficial/probiotic bacteria, including Akkermansia muciniphila, result in reduced inflammation, favoring metabolic and immune health. Immunosuppression is a common feature in conditions of chronic inflammation, and dendritic cells (DCs) represent key targets given their ability to shift the balance toward immunity or tolerance. In this study, the effects of low concentrations of LPS from pathogenic (Escherichia coli and Salmonella enterica) and probiotic (Akkermansia muciniphila) bacterial species on human DC generation and functions were compared. We report that monocyte precursor priming with Escherichia coli and Salmonella enterica LPS forces the differentiation of PD-L1-expressing DCs, releasing high levels of IL-6 and IL-10, and impairs their capacity to drive full TCR-Vδ2 T cell activation. Conversely, comparable concentrations of Akkermansia muciniphila promoted the generation of DCs with preserved activating potential and immunostimulatory properties. These results shed light on potential mechanisms underlying the impact of low endotoxemia on disease risk and pathogenesis, and increase our understanding of the immunomodulatory effects of Akkermansia muciniphila.
Collapse
Affiliation(s)
- Veronica Fertitta
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.F.); (P.F.)
| | - Barbara Varano
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.V.); (M.D.C.)
| | - Manuela Del Cornò
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.V.); (M.D.C.)
| | - Paola Fortini
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.F.); (P.F.)
| | - Anna Aureli
- Institute of Translational Pharmacology, National Research Council, 67100 L’Aquila, Italy;
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.V.); (M.D.C.)
| |
Collapse
|
2
|
Sarajlic M, Neuper T, Vetter J, Schaller S, Klicznik MM, Gratz IK, Wessler S, Posselt G, Horejs-Hoeck J. H. pylori modulates DC functions via T4SS/TNFα/p38-dependent SOCS3 expression. Cell Commun Signal 2020; 18:160. [PMID: 33023610 PMCID: PMC7541176 DOI: 10.1186/s12964-020-00655-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022] Open
Abstract
Background Helicobacter pylori (H. pylori) is a gram-negative bacterium that chronically infects approximately 50% of the world’s human population. While in most cases the infection remains asymptomatic, 10% of infected individuals develop gastric pathologies and 1–3% progress to gastric cancer. Although H. pylori induces severe inflammatory responses, the host’s immune system fails to clear the pathogen and H. pylori can persist in the human stomach for decades. As suppressor of cytokine signaling (SOCS) proteins are important feedback regulators limiting inflammatory responses, we hypothesized that H. pylori could modulate the host’s immune responses by inducing SOCS expression. Methods The phenotype of human monocyte-derived DCs (moDCs) infected with H. pylori was analyzed by flow cytometry and multiplex technology. SOCS expression levels were monitored by qPCR and signaling studies were conducted by means of Western blot. For functional studies, RNA interference-based silencing of SOCS1–3 and co-cultures with CD4+ T cells were performed. Results We show that H. pylori positive gastritis patients express significantly higher SOCS3, but not SOCS1 and SOCS2, levels compared to H. pylori negative patients. Moreover, infection of human moDCs with H. pylori rapidly induces SOCS3 expression, which requires the type IV secretion system (T4SS), release of TNFα, and signaling via the MAP kinase p38, but appears to be independent of TLR2, TLR4, MEK1/2 and STAT proteins. Silencing of SOCS3 expression in moDCs prior to H. pylori infection resulted in increased release of both pro- and anti-inflammatory cytokines, upregulation of PD-L1, and decreased T-cell proliferation. Conclusions This study shows that H. pylori induces SOCS3 via an autocrine loop involving the T4SS and TNFα and p38 signaling. Moreover, we demonstrate that high levels of SOCS3 in DCs dampen PD-L1 expression on DCs, which in turn drives T-cell proliferation. Video Abstract
Collapse
Affiliation(s)
- Muamera Sarajlic
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Theresa Neuper
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Julia Vetter
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg im Muehlkreis, Austria
| | - Susanne Schaller
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg im Muehlkreis, Austria
| | - Maria M Klicznik
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Iris K Gratz
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Silja Wessler
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Gernot Posselt
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria.
| |
Collapse
|
3
|
He M, Chen X, Luo M, Ouyang L, Xie L, Huang Z, Liu A. Suppressor of cytokine signaling 1 inhibits the maturation of dendritic cells involving the nuclear factor kappa B signaling pathway in the glioma microenvironment. Clin Exp Immunol 2020; 202:47-59. [PMID: 32516488 DOI: 10.1111/cei.13476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/22/2020] [Accepted: 05/24/2020] [Indexed: 12/30/2022] Open
Abstract
Recurrence and diffuse infiltration challenge traditional therapeutic strategies for malignant glioma. Immunotherapy appears to be a promising approach to obtain long-term survival. Dendritic cells (DCs), the most specialized and potent antigen-presenting cells (APCs), play an important part in initiating and amplifying both the innate and adaptive immune responses against cancer cells. However, cancer cells can escape from immune surveillance by inhibiting maturation of DCs. Until the present, molecular mechanisms of maturation inhibition of DCs in the tumor microenvironment (TME) have not been fully revealed. Our study showed that pretreatment with tumor-conditioned medium (TCM) collected from supernatant of primary glioma cells significantly suppressed the maturation of DCs. TCM pretreatment significantly changed the morphology of DCs, TCM decreased the expression levels of CD80, CD83, CD86 and interleukin (IL)-12p70, while it increased the expression levels of IL-10, transforming growth factor (TGF)-β and IL-6. RNA-Seq showed that TCM pretreatment significantly increased the gene expression level of suppressor of cytokine signaling 1 (SOCS1) in DCs. suppressor of cytokine signaling 1 (SOCS1) knock-down significantly antagonized the maturation inhibition of DCs by TCM, which was demonstrated by the restoration of maturation markers. TCM pretreatment also significantly suppressed T cell viability and T helper type 1 (Th1) response, and SOCS1 knock-down significantly antagonized this suppressive effect. Further, TCM pretreatment significantly suppressed p65 nuclear translocation and transcriptional activity in DCs, and SOCS1 knock-down significantly attenuated this suppressive effect. In conclusion, our research demonstrates that TCM up-regulate SOCS1 to suppress the maturation of DCs via the nuclear factor-kappa signaling pathway.
Collapse
Affiliation(s)
- M He
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - X Chen
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - M Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - L Ouyang
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - L Xie
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Z Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Neurosurgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - A Liu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
4
|
Hildebrand D, Metz-Zumaran C, Jaschkowitz G, Heeg K. Silencing SOCS1 via Liposome-Packed siRNA Sustains TLR4-Ligand Adjuvant. Front Immunol 2019; 10:1279. [PMID: 31214204 PMCID: PMC6558036 DOI: 10.3389/fimmu.2019.01279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 05/20/2019] [Indexed: 11/13/2022] Open
Abstract
Infectious diseases remain one of the leading causes of death worldwide. Vaccination is a powerful instrument to avert a variety of those by inducing a pathogen-specific immune response and ensure a long-lasting protection against the respective infection. Nevertheless, due to increasing numbers of immunocompromised patients and emergence of more aggressive pathogens existing vaccination techniques are limited. In our study we investigated a new strategy to strengthen vaccine adjuvant in order to increase immunity against infectious diseases. The strategy is based on an amplification of Toll-like receptor 4 (TLR4) -induced activation of antigen-presenting cells (APCs) by turning off a powerful endogenous inhibitor of APC-activation. TLR4 signaling induces the release of cytokines that bind autocrine and paracrine to receptors, activating the Janus kinase (JAK) 2/signal transducers and activators of transcription (STAT) 3 cascade. Subsequently, STAT3 induces expression of suppressor of cytokine signaling (SOCS) 1 that terminates the inflammatory response. In the approach, TLR4-adjuvant monophosphoryl lipid A (MPLA)-stimulated monocyte-activation is reinforced and sustained by silencing SOCS1 via lipid nanoparticle-enclosed siRNA (L-siRNA). L-siRNA is transported into primary cells without any toxic side effects and protected from early degradation. Through lipid core-embedded functional groups the lipid particle escapes from endosomes and releases the siRNA when translocated into the cytoplasm. SOCS1 is potently silenced, and SOCS1-mediated termination of NFκB signaling is abrogated. Consequently, the MPLA-stimulated activation of APCs, monitored by release of pro-inflammatory cytokines such as IL-6, TNFα, and IL-1β, upregulation of MHC class II molecules and costimulatory CD80/CD86 is strongly enhanced and prolonged. SOCS1-silenced APCs, pulsed with liposomal tetanus light chain toxin (TeTxLC) antigen, activate autologous T cells much more intensively than SOCS1-expressing cells. Importantly, expansion of cocultured CD4+ as well as CD8+ T cells is remarkably enhanced. Furthermore, our results point toward a broad T helper cell response as TH1 typical as well as TH2 characteristic cytokines are elevated. Taken together, this study in the human system comprises a translational potential to develop more effective vaccines against infectious diseases by inhibition of the endogenous negative-feedback loop in APCs.
Collapse
Affiliation(s)
- Dagmar Hildebrand
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany.,DZIF German Center for Infection Research, Braunschweig, Germany
| | - Camila Metz-Zumaran
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany.,DZIF German Center for Infection Research, Braunschweig, Germany
| | - Greta Jaschkowitz
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Heeg
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany.,DZIF German Center for Infection Research, Braunschweig, Germany
| |
Collapse
|
5
|
Gargiulo E, Paggetti J, Moussay E. Hematological Malignancy-Derived Small Extracellular Vesicles and Tumor Microenvironment: The Art of Turning Foes into Friends. Cells 2019; 8:cells8050511. [PMID: 31137912 PMCID: PMC6562645 DOI: 10.3390/cells8050511] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 02/07/2023] Open
Abstract
Small extracellular vesicles (small EVs) are commonly released by all cells, and are found in all body fluids. They are implicated in cell to cell short- and long-distance communication through the transfer of genetic material and proteins, as well as interactions between target cell membrane receptors and ligands anchored on small EV membrane. Beyond their canonical functions in healthy tissues, small EVs are strategically used by tumors to communicate with the cellular microenvironment and to establish a proper niche which would ultimately allow cancer cell proliferation, escape from the immune surveillance, and metastasis formation. In this review, we highlight the effects of hematological malignancy-derived small EVs on immune and stromal cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Ernesto Gargiulo
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, 84, val fleuri, L-1526 Luxembourg, Luxembourg.
| | - Jerome Paggetti
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, 84, val fleuri, L-1526 Luxembourg, Luxembourg.
| | - Etienne Moussay
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, 84, val fleuri, L-1526 Luxembourg, Luxembourg.
| |
Collapse
|
6
|
Iliev DB, Lagos L, Thim HL, Jørgensen SM, Krasnov A, Jørgensen JB. CpGs Induce Differentiation of Atlantic Salmon Mononuclear Phagocytes Into Cells With Dendritic Morphology and a Proinflammatory Transcriptional Profile but an Exhausted Allostimulatory Activity. Front Immunol 2019; 10:378. [PMID: 30918507 PMCID: PMC6424866 DOI: 10.3389/fimmu.2019.00378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Abstract
Due to their ability to present foreign antigens and prime naïve T cells, macrophages, and dendritic cells (DCs) are referred to as professional antigen-presenting cells (APCs). Although activated macrophages may function as APCs, these cells are particularly effective at directly engaging pathogens through phagocytosis, and production of antimicrobial compounds. On the other hand, DCs possess superb antigen-presenting and costimulatory capacity and they are essential for commencement and regulation of adaptive immune responses. In in vitro models, development of mature mammalian DCs from monocytes requires sequential exposure to growth factors (including GM-CSF and IL-4) and proinflammatory stimuli such as toll-like receptor (TLR) ligands. Currently, except for IL-4/13, neither orthologs nor functional analogs of the growth factors which are essential for the differentiation of mammalian DCs (including GM-CSF and FLT3) have been identified in teleosts and data about differentiation of piscine APCs is scant. In the present study, primary salmon mononuclear phagocytes (MPs) stimulated in vitro for 5-7 days with a B-class CpG oligodeoxynucleotides (ODN 2006PS) underwent morphological differentiation and developed "dendritic" morphology, characterized by long, branching pseudopodia. Transcriptional profiling showed that these cells expressed high levels of proinflammatory mediators characteristic for M1 polarized MPs. However, the cells treated with CpGs for 7 days downregulated their surface MHCII molecules as well as their capacity to endocytose ovalbumin and exhibited attenuated allostimulatory activity. This concurred with transcriptional downregulation of costimulatory CD80/86 and upregulation of inhibitory CD274 (B7-H1) genes. Despite their exhausted allostimulatory activity, these cells were still responsive to re-stimulation with gardiquimod (a TLR7/8 ligand) and further upregulated a wide array of immune genes including proinflammatory mediators such as intereukin-1 beta and tumor necrosis factor. Overall, the presented data highlight the disparate effects TLR ligands may have on the proinflammatory status of APCs, on one side, and their antigen-presenting/costimulatory functions, on the other. These findings also indicate that despite the poor phylogenetic conservation of the growth factors involved in the differentiation of DCs, some of the processes that orchestrate the development and the differentiation of professional APCs are conserved between teleosts in mammals.
Collapse
Affiliation(s)
- Dimitar B Iliev
- The Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Gene Regulation, Institute of Molecular Biology 'Roumen Tsanev', Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Leidy Lagos
- The Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Hanna L Thim
- The Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | | | | | - Jorunn B Jørgensen
- The Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
7
|
Su H, Peng B, Zhang Z, Liu Z, Zhang Z. The Mycobacterium tuberculosis glycoprotein Rv1016c protein inhibits dendritic cell maturation, and impairs Th1 /Th17 responses during mycobacteria infection. Mol Immunol 2019; 109:58-70. [PMID: 30856410 DOI: 10.1016/j.molimm.2019.02.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/13/2019] [Accepted: 02/27/2019] [Indexed: 12/20/2022]
Abstract
The myobacterial factors and the associated mechanism by which Mycobacterium tuberculosis (Mtb) evades the host immune surveillance system remain widely unexplored. Here, we found that overexpressing Rv1016c, a mannosylated protein of M. tuberculosis in BCG (rBCG-Rv1016c) led to increased virulence of the recombined BCG in the severe-combined immunodeficient (SCID) mice model and to a loss of protective efficacy in a zebrafish-M. marinum model, compared to wild type BCG. Further investigations on the effects of rBCG-Rv1016c on the host innate immunity revealed that rBCG-Rv1016c decreased the production of cytokines IL-2, IL-12p70, TGF-β, IL-6 as well as of the co-stimulatory molecules CD80, CD86, MHC-I and MHC-II by the infected DCs. These effects were mimicked by rBCG-Rv1016cHis, which carried an extra 6-His tag at the C-terminus of Rv1016c. Relatively to BCG infected DCs, the rBCG-Rv1016c-infected DCs failed to polarize naïve T cells to Th1- and Th17-type cells to secret IFN-γ and IL-17. Additionally, T lymphocytes from BCG- infected mice showed significantly less proliferation and production of IFN-γ and IL-17. Similarly, rBCG-Rv1016c mice released a higher level of IL-10 in response to rBCG-Rv1016c stimulation than wild type BCG infected mice. Furthermore, DCs from TLR-2 knockout mice showed no reduction in IL-6, IL-12 p70 and TGF-β secretion in response to rBCG-Rv1016c infection, compared to DCs infected with BCG. We propose that Rv1016c interferes in differentiation of the DCs by targeting suppressor of cytokine signaling (SOCS) 1 and SOCS3 expression, which subsequently leads to the reduction in STAT-1 and STAT-6 phosphorylation. These findings open new perspectives regarding the immunosuppressive strategies adopted by Mtb to survive in the host.
Collapse
Affiliation(s)
- Haibo Su
- GMU-GIBH Joint School of Life Science, Guangzhou Medical University, No. 195 Dongfengxi Road, Guangzhou, 510000, China; Guangdong Second Provincial General Hospital, No. 466 Xingang Road, Guangzhou, 510220, China
| | - Baozhou Peng
- GMU-GIBH Joint School of Life Science, Guangzhou Medical University, No. 195 Dongfengxi Road, Guangzhou, 510000, China
| | - Zhen Zhang
- Guangdong Second Provincial General Hospital, No. 466 Xingang Road, Guangzhou, 510220, China
| | - Zijian Liu
- GMU-GIBH Joint School of Life Science, Guangzhou Medical University, No. 195 Dongfengxi Road, Guangzhou, 510000, China
| | - Zhi Zhang
- Guangdong Second Provincial General Hospital, No. 466 Xingang Road, Guangzhou, 510220, China.
| |
Collapse
|
8
|
Perdijk O, van Neerven RJJ, van den Brink E, Savelkoul HFJ, Brugman S. The oligosaccharides 6'-sialyllactose, 2'-fucosyllactose or galactooligosaccharides do not directly modulate human dendritic cell differentiation or maturation. PLoS One 2018; 13:e0200356. [PMID: 29990329 PMCID: PMC6039038 DOI: 10.1371/journal.pone.0200356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/25/2018] [Indexed: 01/25/2023] Open
Abstract
Breast milk plays an important role in immune development in early life and protects against diseases later in life. A wide range of the beneficial effects of breast milk are attributed to human milk oligosaccharides (HMOs) as well as components such as vitamin D3 (VitD3) or TGFβ. One mechanism by which HMOs might contribute to immune homeostasis and protection against disease is the induction of a local tolerogenic milieu. In this study we investigated the effect of the HMOs 6’-sialyllactose (6’SL) and 2’-fucosyllactose (2’FL) as well as prebiotic galactooligosaccharides (GOS) on DC differentiation and maturation. Isolated CD14+ monocytes were cultured for six days in the presence of GM-CSF and IL-4 with or without 6’SL, 2’FL, GOS, VitD3 or TGFβ. Additionally, immature VitD3DC, TGFβDC and moDC were used as different DC types to investigate the effect of 6’SL, 2’FL and GOS on DC maturation. Surface marker expression and cytokine production was measured by flow cytometry and cytometric bead array, respectively. Unlike TGFβ and vitD3, the oligosaccharides 6’SL, 2’FL and GOS did not influence DC differentiation. Next, we studied the effect of 6’SL, 2’FL and GOS on maturation of moDC, VitD3DC and TGFβDC that showed different profiles of HMO-binding receptors. 6’SL, 2’FL and GOS did not modulate LPS-induced maturation, even though their putative receptors were present on the different DCs types. Thus, whereas VitD3 and TGFβ halt DC differentiation, which results in phenotypically distinct tolerogenic DCs, 6’SL, 2’FL and GOS do not alter DC differentiation or maturation of in vitro differentiated DC types.
Collapse
Affiliation(s)
- Olaf Perdijk
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
| | - R. J. Joost van Neerven
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
- FrieslandCampina, Amersfoort, the Netherlands
| | - Erik van den Brink
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology group, Wageningen University, Wageningen, the Netherlands
- * E-mail:
| |
Collapse
|
9
|
Serum amyloid A inhibits dendritic cell differentiation by suppressing GM-CSF receptor expression and signaling. Exp Mol Med 2017; 49:e369. [PMID: 28857084 PMCID: PMC5579511 DOI: 10.1038/emm.2017.120] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/18/2022] Open
Abstract
In this study, we report that an acute phase reactant, serum amyloid A (SAA), strongly inhibits dendritic cell differentiation induced by GM-CSF plus IL-4. SAA markedly decreased the expression of MHCII and CD11c. Moreover, SAA decreased cell surface GM-CSF receptor expression. SAA also decreased the expression of PU.1 and C/EBPα, which play roles in the expression of GM-CSF receptor. This inhibitory response by SAA is partly mediated by the well-known SAA receptors, Toll-like receptor 2 and formyl peptide receptor 2. Taken together, we suggest a novel insight into the inhibitory role of SAA in dendritic cell differentiation.
Collapse
|
10
|
Amir M, Aqdas M, Nadeem S, Siddiqui KF, Khan N, Sheikh JA, Agrewala JN. Diametric Role of the Latency-Associated Protein Acr1 of Mycobacterium tuberculosis in Modulating the Functionality of Pre- and Post-maturational Stages of Dendritic Cells. Front Immunol 2017; 8:624. [PMID: 28611779 PMCID: PMC5447689 DOI: 10.3389/fimmu.2017.00624] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
It is instrumental for the Mycobacterium tuberculosis (Mtb) to persist within its host in dormancy. Mtb represses most of its metabolic machinery during latency, but upregulates the expression of latency-associated protein alpha-crystallin protein (Acr1). Therefore, it is imperative to understand how throughout dormancy, Mtb employs Acr1 to regulate the host immunity. This study reveals that Acr1 exhibits divergent effect on the pre- and post-maturation stages of dendritic cells (DCs). In the current study, we demonstrate that early encounter of bone marrow cells with Acr1 while differentiating into DCs (AcrDCpre), leads to impairment in their maturation. In contrast, when exposed to Acr1 after maturation (AcrDCpost), DCs show augmentation in their activity, secretion of TNF-α, IL-12, IL-6, and activation of T cells. Additionally, AcrDCpost promoted the polarization of naïve CD4 T cells to Th1 cells and Th17 cells and restricted the intracellular growth of Mtb. Furthermore, these DCs upregulated the expression of CCR7 and exhibited enhanced migratory capabilities. The discrete impact of Acr1 on DCs is mediated through a mechanism involving STAT-1, SOCS-3, ERK, TLR-4, and NF-κB signaling pathways. This study reveals the unprecedented role of Acr1 in distinctly modulating the function of DCs at different stages of maturation.
Collapse
Affiliation(s)
- Mohammed Amir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sajid Nadeem
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Kaneez F Siddiqui
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Nargis Khan
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javaid A Sheikh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
11
|
Dendritic cells that highly express SOCS1 induce T-cell hypo-responsiveness and prolong islet allograft survival. Cell Immunol 2017; 314:36-41. [DOI: 10.1016/j.cellimm.2017.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 01/07/2017] [Accepted: 01/27/2017] [Indexed: 11/17/2022]
|
12
|
Ilangumaran S, Bobbala D, Ramanathan S. SOCS1: Regulator of T Cells in Autoimmunity and Cancer. Curr Top Microbiol Immunol 2017; 410:159-189. [PMID: 28900678 DOI: 10.1007/82_2017_63] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SOCS1 is a negative feedback regulator of cytokine and growth factor receptor signaling, and plays an indispensable role in attenuating interferon gamma signaling. Studies on SOCS1-deficient mice have established a crucial role for SOCS1 in regulating CD8+ T cell homeostasis. In the thymus, SOCS1 prevents thymocytes that had failed positive selection from surviving and expanding, ensures negative selection and prevents inappropriate developmental skewing toward the CD8 lineage. In the periphery, SOCS1 not only controls production of T cell stimulatory cytokines but also attenuates the sensitivity of CD8+ T cells to synergistic cytokine stimulation and antigen non-specific activation. As cytokine stimulation of CD8+ T lymphocytes increases their sensitivity to low affinity TCR ligands, SOCS1 likely contributes to peripheral T cell tolerance by putting brakes on aberrant T cell activation driven by inflammatory cytokines. In addition, SOCS1 is critical to maintain the stability of T regulatory cells and control their plasticity to become pathogenic Th17 and Th1 cells under the harmful influence of inflammatory cytokines. SOCS1 also regulates T cell activation by dendritic cells via modulating their generation, maturation, antigen presentation, costimulatory signaling, and cytokine production. The above control mechanisms of SOCS1 on T cells, T regulatory cells and dendritic cells collectively contribute to immunological tolerance and prevent autoimmune manifestation. On other hand, silencing SOCS1 in dendritic cells or CD8+ T cells stimulates efficient antitumor immunity. Thus, even though SOCS1 is not a cell surface checkpoint inhibitor, its regulatory functions on T cell responses qualify SOCS1as a "non-classical" checkpoint blocker. SOCS1 also functions as a tumor suppressor in cancer cells by regulating oncogenic signal transduction pathways. The loss of SOCS1 expression observed in many tumors may have an impact on classical checkpoint pathways. The potential to exploit SOCS1 to treat inflammatory/autoimmune diseases and elicit antitumor immunity is discussed.
Collapse
Affiliation(s)
- Subburaj Ilangumaran
- Immunology Division, Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada.
| | - Diwakar Bobbala
- Immunology Division, Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Sheela Ramanathan
- Immunology Division, Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| |
Collapse
|
13
|
Zhao HM, Xu R, Huang XY, Cheng SM, Huang MF, Yue HY, Wang X, Zou Y, Lu AP, Liu DY. Curcumin Suppressed Activation of Dendritic Cells via JAK/STAT/SOCS Signal in Mice with Experimental Colitis. Front Pharmacol 2016; 7:455. [PMID: 27932984 PMCID: PMC5122716 DOI: 10.3389/fphar.2016.00455] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) play a pivotal role as initiators in the pathogenesis of inflammatory bowel disease and are regulated by the JAK/STAT/SOCS signaling pathway. As a potent anti-inflammatory compound, curcumin represents a viable treatment alternative or adjunctive therapy in the management of chronic inflammatory bowel disease (IBD). The mechanism of curcumin treated IBD on DCs is not completely understood. In the present study, we explored the mechanism of curcumin treated experimental colitis by observing activation of DCs via JAK/STAT/SOCS signaling pathway in colitis mice. Experimental colitis was induced by 2, 4, 6-trinitrobenzene sulfonic acid. After 7 days treatment with curcumin, its therapeutic effect was verified by decreased colonic weight, histological scores, and remitting pathological injury. Meanwhile, the levels of major histocompatibility complex class II and DC costimulatory molecules (CD83, CD28, B7-DC, CD40, CD40 L, and TLR2) were inhibited and followed the up-regulated levels of IL-4, IL-10, and IFN-γ, and down-regulated GM-CSF, IL-12p70, IL-15, IL-23, and TGF-β1. A key finding was that the phosphorylation of the three members (JAK2, STAT3, and STAT6) of the JAK/STAT/SOCS signaling pathway was inhibited, and the three downstream proteins (SOCS1, SOCS3, and PIAS3) from this pathway were highly expressed. In conclusion, curcumin suppressed the activation of DCs by modulating the JAK/STAT/SOCS signaling pathway to restore immunologic balance to effectively treat experimental colitis.
Collapse
Affiliation(s)
- Hai-Mei Zhao
- School of Basic Medical Sciences, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Rong Xu
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Xiao-Ying Huang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Shao-Min Cheng
- School of Basic Medical Sciences, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Min-Fang Huang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Hai-Yang Yue
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Xin Wang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Yong Zou
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| | - Ai-Ping Lu
- School of Chinese Medicine, Hong Kong Baptist University Kowloon Tong, China
| | - Duan-Yong Liu
- Science and Technology College, Jiangxi University of Traditional Chinese Medicine Nanchang, China
| |
Collapse
|
14
|
Motran CC, Ambrosio LF, Volpini X, Celias DP, Cervi L. Dendritic cells and parasites: from recognition and activation to immune response instruction. Semin Immunopathol 2016; 39:199-213. [DOI: 10.1007/s00281-016-0588-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/22/2016] [Indexed: 12/20/2022]
|
15
|
Sachdeva M, Sharma A, Arora SK. Functional Impairment of Myeloid Dendritic Cells during Advanced Stage of HIV-1 Infection: Role of Factors Regulating Cytokine Signaling. PLoS One 2015; 10:e0140852. [PMID: 26492336 PMCID: PMC4619614 DOI: 10.1371/journal.pone.0140852] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/01/2015] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Severely immunocompromised state during advanced stage of HIV-1 infection has been linked to functionally defective antigen presentation by dendritic cells (DCs). The molecular mechanisms behind DC impairment are still obscure. We investigated changes in DC function and association of key regulators of cytokine signaling during different stages of HIV-1 infection and following antiretroviral therapy (ART). METHODS Phenotypic and functional characteristics of circulating myeloid DCs (mDCs) in 56 ART-naive patients (23 in early and 33 in advanced stage of disease), 36 on ART and 24 healthy controls were evaluated. Sixteen patients were studied longitudinally prior-to and 6 months after the start of ART. For functional studies, monocyte-derived DCs (Mo-DCs) were evaluated for endocytosis, allo-stimulation and cytokine secretion. The expression of suppressor of cytokine signaling (SOCS)-1 and other regulators of cytokine signaling was evaluated by real-time RT-PCR. RESULTS The ability to respond to an antigenic stimulation was severely impaired in patients in advanced HIV-1 disease which showed partial recovery in the treated group. Mo-DCs from patients with advanced HIV-disease remained immature with low allo-stimulation and reduced cytokine secretion even after TLR-4 mediated stimulation ex-vivo. The cells had an increased expression of negative regulatory factors like SOCS-1, SOCS-3, SH2-containing phosphatase (SHP)-1 and a reduced expression of positive regulators like Janus kinase (JAK)2 and Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)1. A functional recovery after siRNA mediated silencing of SOCS-1 in these mo-DCs confirms the role of negative regulatory factors in functional impairment of these cells. CONCLUSIONS Functionally defective DCs in advanced stage of HIV-1 infection seems to be due to imbalanced state of negative and positive regulatory gene expression. Whether this is a cause or effect of increased viral replication at this stage of disease, needs further investigation. The information may be useful in design of novel therapeutic targets for better management of disease.
Collapse
Affiliation(s)
- Meenakshi Sachdeva
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Aman Sharma
- Department of Internal Medicine, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Sunil K. Arora
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
- * E-mail:
| |
Collapse
|
16
|
Kim SK, Yun CH, Han SH. Dendritic Cells Differentiated from Human Umbilical Cord Blood-Derived Monocytes Exhibit Tolerogenic Characteristics. Stem Cells Dev 2015. [PMID: 26203805 DOI: 10.1089/scd.2014.0600] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human umbilical cord blood (UCB) is rich in diverse hematopoietic stem cells that are competent to differentiate into various cell types with immunological compatibility at transplantation. Thus, UCB is a potential source for the preparation of dendritic cells (DCs) to be used for cell therapy against inflammatory disorders or cancers. However, the immunological properties of UCB-derived DCs are not fully characterized. In this study, we investigated the phenotypes and functions of UCB monocyte-derived DCs (UCB-DCs) in comparison with those of adult peripheral blood (APB) monocyte-derived DCs (APB-DCs). UCB-DCs contained less CD1a(+) DCs, which is known as immunostimulatory DCs, than APB-DCs. UCB-DCs exhibited lower expression of CD80, MHC proteins, and DC-SIGN, but higher endocytic activity, than APB-DCs. Lipopolysaccharide stimulation of UCB-DCs minimally augmented the expression of maturation markers and production of interleukin (IL)-12 and tumor necrosis factor (TNF)-α, but potently expressed IL-10. When UCB-DCs were cocultured with CD14(+) cell-depleted allogeneic peripheral blood mononuclear cells, they weakly induced the proliferation, surface expression of activation markers, and interferon (IFN)-γ production of T lymphocytes compared with APB-DCs. UCB possessed higher levels of prostaglandin E2 (PGE2) than APB, which might be responsible for tolerogenic phenotypes and functions of UCB-DCs. Indeed, APB-DCs prepared in the presence of PGE2 exhibited CD1a(-)CD14(+) phenotypes with tolerogenic properties, including weak maturation, impaired IL-12 production, and negligible T lymphocyte activation as UCB-DCs did. Taken together, we suggest that UCB-DCs have tolerogenic properties, which might be due to PGE2 highly sustained in UCB.
Collapse
Affiliation(s)
- Sun Kyung Kim
- 1 Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University , Seoul, Republic of Korea
| | - Cheol-Heui Yun
- 2 Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University , Seoul, Republic of Korea
| | - Seung Hyun Han
- 1 Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University , Seoul, Republic of Korea
| |
Collapse
|
17
|
Patni S, Bryant AH, Wynen LP, Seager AL, Morgan G, Thornton CA. Functional activity but not gene expression of toll-like receptors is decreased in the preterm versus term human placenta. Placenta 2015; 36:1031-8. [PMID: 26190036 DOI: 10.1016/j.placenta.2015.06.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Toll-like receptor (TLR) activity within gestation-associated tissues might have a role in normal pregnancy progression as well as adverse obstetric outcomes such as preterm birth (PTB). METHODS The expression and activity of TLRs 1-9 in placentas collected following preterm vaginal delivery after infection-associated preterm labour (IA-PTL) at 25-36 weeks of gestation (preterm-svd, n = 10) were compared with those obtained after normal vaginal delivery at term (term-laboured; n = 17). Placental explants were cultured in the presence of agonists for TLR2, 3, 4, 5, 7, 8 and 9 and cytokine production after 24 h examined. Expression of TLR transcripts was determined using real time quantitative PCR. RESULTS Reactivity to all agonists except CpG oligonucleotides was observed indicating that other than TLR9 all of the receptors studied yielded functional responses both term and preterm. Significantly less TNFα and IL-6, but not IL-10, were produced by preterm than term samples in response to all TLR agonists. Changes in TLR mRNA expression did not underlie functional differences in the preterm and term groups; nor does a pre-exposure/tolerance model mimic this finding. While glucocorticoids suppressed cytokine production in an in vitro model using term tissue the association between lower gestational age and decreased cytokine outputs suggests a temporally regulated response. DISCUSSION Pro-inflammatory cytokine output in response to multiple TLR ligands was decreased in the preterm compared to the term placenta but gene expression for each TLR tended to be similar. Reduced cytokine production by the preterm placenta in response to stimulation of TLRs therefore must be regulated at the post-transcriptional level in a gestational age dependent manner.
Collapse
Affiliation(s)
- Shalini Patni
- Princess of Wales Maternity Unit, Birmingham Heartlands Hospital, Birmingham, UK
| | - Aled H Bryant
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Louise P Wynen
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Anna L Seager
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Gareth Morgan
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Catherine A Thornton
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, UK.
| |
Collapse
|
18
|
Lopez de Lapuente A, Pinto-Medel MJ, Astobiza I, Alloza I, Comabella M, Malhotra S, Montalban X, Zettl UK, Rodríguez-Antigüedad A, Fernández O, Vandenbroeck K. Cell-specific effects in different immune subsets associated with SOCS1 genotypes in multiple sclerosis. Mult Scler 2015; 21:1498-512. [PMID: 25623250 DOI: 10.1177/1352458514566418] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/19/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) near SOCS1 are associated with multiple sclerosis (MS), but the most important SNPs in the area and mechanisms by which they influence the disease are unknown. METHODS A haplotype-tagging association study was performed covering 60.5kbp around SOCS1, and the index SNP was validated in a total of 2292 individuals. mRNA expression of SOCS1 and nearby genes was measured in MS patients with different disease courses and healthy controls. SOCS1 protein expression was studied by flow cytometry in a separate cohort of patients and controls. Differentiation and maturation of monocyte-derived dendritic cells (moDCs) were also studied. RESULTS One SNP, rs423674, reached genome-wide significance. No genotype-specific mRNA expression differences were seen, but, by flow cytometry, significant interactions were observed between genotypes for rs423674 and disease activity (relapse or remission) in B cells and regulatory T cells. Furthermore, homozygotes for the risk allele (GG) showed higher levels of CD1a and CD86 than carriers of the protective allele (GT) in immature moDCs and a greater increase of HLA-DR+ cell percentage than GT heterozygotes upon maturation. CONCLUSIONS rs423674, or its genetic proxies, may influence MS risk by modulating SOCS1 expression in a cell-specific manner and by influencing dendritic cell function.
Collapse
Affiliation(s)
- Aitzkoa Lopez de Lapuente
- Neurogenomiks Group, Universidad del País Vasco (UPV/EHU), Spain/Achucarro Basque Center for Neuroscience, Spain
| | - María Jesús Pinto-Medel
- Research Laboratory, Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya and Fundación IMABIS
| | - Ianire Astobiza
- Neurogenomiks Group, Universidad del País Vasco (UPV/EHU), Spain/Achucarro Basque Center for Neuroscience, Spain
| | - Iraide Alloza
- Neurogenomiks Group, Universidad del País Vasco (UPV/EHU), Spain/ Achucarro Basque Center for Neuroscience, Spain/ IKERBASQUE, Basque Foundation for Science, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain
| | - Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain
| | - Uwe K Zettl
- University of Rostock, Department of Neurology, Germany
| | | | - Oscar Fernández
- Research Laboratory, Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya and Fundación IMABIS
| | - Koen Vandenbroeck
- Neurogenomiks Group, Universidad del País Vasco (UPV/EHU), Spain/ Achucarro Basque Center for Neuroscience, Spain/ IKERBASQUE, Basque Foundation for Science, Spain
| |
Collapse
|
19
|
Rizzo M, Bayo J, Piccioni F, Malvicini M, Fiore E, Peixoto E, García MG, Aquino JB, Gonzalez Campaña A, Podestá G, Terres M, Andriani O, Alaniz L, Mazzolini G. Low molecular weight hyaluronan-pulsed human dendritic cells showed increased migration capacity and induced resistance to tumor chemoattraction. PLoS One 2014; 9:e107944. [PMID: 25238610 PMCID: PMC4169605 DOI: 10.1371/journal.pone.0107944] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 08/18/2014] [Indexed: 12/12/2022] Open
Abstract
We have shown that ex vivo pre-conditioning of bone marrow-derived dendritic cells (DC) with low molecular weight hyaluronan (LMW HA) induces antitumor immunity against colorectal carcinoma (CRC) in mice. In the present study we investigated the effects of LMW HA priming on human-tumor-pulsed monocytes-derived dendritic cells (DC/TL) obtained from healthy donors and patients with CRC. LMW HA treatment resulted in an improved maturation state of DC/TL and an enhanced mixed leucocyte reaction activity in vivo. Importantly, pre-conditioning of DC/TL with LMW HA increased their ability to migrate and reduced their attraction to human tumor derived supernatants. These effects were associated with increased CCR7 expression levels in DC. Indeed, a significant increase in migratory response toward CCL21 was observed in LMW HA primed tumor-pulsed monocyte-derived dendritic cells (DC/TL/LMW HA) when compared to LWM HA untreated cells (DC/TL). Moreover, LMW HA priming modulated other mechanisms implicated in DC migration toward lymph nodes such as the metalloproteinase activity. Furthermore, it also resulted in a significant reduction in DC migratory capacity toward tumor supernatant and IL8 in vitro. Consistently, LMW HA dramatically enhanced in vivo DC recruitment to tumor-regional lymph nodes and reduced DC migration toward tumor tissue. This study shows that LMW HA--a poorly immunogenic molecule--represents a promising candidate to improve human DC maturation protocols in the context of DC-based vaccines development, due to its ability to enhance their immunogenic properties as well as their migratory capacity toward lymph nodes instead of tumors.
Collapse
Affiliation(s)
- Manglio Rizzo
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Flavia Piccioni
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Mariana Malvicini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), CABA, Buenos Aires, Argentina
| | - Esteban Fiore
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Estanislao Peixoto
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Mariana G. García
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), CABA, Buenos Aires, Argentina
| | - Jorge B. Aquino
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), CABA, Buenos Aires, Argentina
| | - Ariel Gonzalez Campaña
- Department of Surgery, Hospital Austral, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Gustavo Podestá
- Department of Surgery, Hospital Austral, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Marcelo Terres
- Department of Surgery, Hospital Austral, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Oscar Andriani
- Department of Surgery, Hospital Austral, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Laura Alaniz
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), CABA, Buenos Aires, Argentina
- CIT NOBA, Universidad Nacional del Noroeste de la Pcia de Bs. As (UNNOBA), Junín, Buenos Aires, Argentina
- * E-mail: (GM); (LA)
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), CABA, Buenos Aires, Argentina
- * E-mail: (GM); (LA)
| |
Collapse
|
20
|
Siddiqui KF, Amir M, Gurram RK, Khan N, Arora A, Rajagopal K, Agrewala JN. Latency-associated protein Acr1 impairs dendritic cell maturation and functionality: a possible mechanism of immune evasion by Mycobacterium tuberculosis. J Infect Dis 2013; 209:1436-45. [PMID: 24218502 DOI: 10.1093/infdis/jit595] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mycobacterium tuberculosis (M. tuberculosis) in latently infected individuals survives and thwarts the attempts of eradication by the immune system. During latency, Acr1 is predominantly expressed by the bacterium. However, whether M. tuberculosis exploits its Acr1 in impairing the host immunity remains widely unexplored. Hence, currently we have investigated the role of Acr1 in influencing the differentiation and function of dendritic cells (DCs), which play a cardinal role in innate and adaptive immunity. Therefore, for the first time, we have revealed a novel mechanism of mycobacterial Acr1 in inhibiting the maturation and differentiation of DCs by inducing tolerogenic phenotype by modulating the expression of PD-L1; Tim-3; indoleamine 2, 3-dioxygenase (IDO); and interleukin 10. Furthermore, Acr1 interferes in the differentiation of DCs by targeting STAT-6 and STAT-3 pathways. Continuous activation of STAT-3 inhibited the translocation of NF-κB in Acr1-treated DCs. Furthermore, Acr1 also augmented the induction of regulatory T cells. These DCs displayed decline in their antigen uptake capacity and reduced ability to help T cells. Interestingly, M. tuberculosis exhibited better survival in Acr1-treated DCs. Thus, this study provides a crucial insight into a strategy adopted by M. tuberculosis to survive in the host by impairing the function of DCs.
Collapse
Affiliation(s)
- Kaneez F Siddiqui
- Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Chandigarh, India
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang Y, Wang S, Ding Y, Ye Y, Xu Y, He H, Li Q, Mi Y, Guo C, Lin Z, Liu T, Zhang Y, Chen Y, Yan J. A suppressor of cytokine signaling 1 antagonist enhances antigen-presenting capacity and tumor cell antigen-specific cytotoxic T lymphocyte responses by human monocyte-derived dendritic cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1449-56. [PMID: 23885028 PMCID: PMC3889590 DOI: 10.1128/cvi.00130-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 07/15/2013] [Indexed: 12/23/2022]
Abstract
The suppressor of cytokine signaling 1 (SOCS1) has emerged as a critical inhibitory molecule for controlling the cytokine response and antigen presentation by dendritic cells (DCs), thereby regulating the magnitude of both innate and adaptive immunity. The aim of this study was to investigate whether the SOCS1 antagonist pJAK2(1001-1013) peptide can weaken or block the inhibition function of SOCS1 in DCs by evaluating the phenotype and cytokine production, antigen-presenting, and specific T-cell-activating capacities of DCs electroporated with human gastric cancer cell total RNA. Furthermore, STAT1 activation of the JAK/STAT signal pathway mediated by SOCS1 was analyzed by Western blotting. The results demonstrate that the SOCS1 antagonist pJAK2(1001-1013) peptide upregulated the expression of the maturation marker (CD83) and costimulatory molecule (CD86) of RNA-electroporated human monocyte-derived mature DCs (mDCs), potentiated the capacity of mDCs to induce T-cell proliferation, stimulated the secretion of proinflammatory cytokines, and enhanced the cytotoxicity of tumor cell antigen-specific CTLs activated by human gastric cancer cell total RNA-electroporated mDCs. Data from Western blot analysis indicate that STAT1 was further activated in pJAK2(1001-1013) peptide-loaded mDCs. These results imply that the SOCS1 antagonist pJAK2(1001-1013) peptide is an effective reagent for the enhancement of antigen-specific antitumor immunity by DCs.
Collapse
Affiliation(s)
- Yongjun Wang
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Shengyu Wang
- Cancer Research Center, Medical College of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yuan Ding
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yanhua Ye
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yingyi Xu
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Huixiang He
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Qiaozhen Li
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yanjun Mi
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Chunhua Guo
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Zhicai Lin
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Tao Liu
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yaya Zhang
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yuqiang Chen
- Department of Oncology, 174th Hospital of the Chinese People's Liberation Army, Affiliated Chenggong Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Jianghua Yan
- Cancer Research Center, Medical College of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| |
Collapse
|
22
|
Fekete T, Szabo A, Beltrame L, Vivar N, Pivarcsi A, Lanyi A, Cavalieri D, Rajnavölgyi E, Rethi B. Constraints for monocyte-derived dendritic cell functions under inflammatory conditions. Eur J Immunol 2011; 42:458-69. [PMID: 22057588 DOI: 10.1002/eji.201141924] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/30/2011] [Accepted: 10/26/2011] [Indexed: 11/12/2022]
Abstract
The activation of TLRs expressed by macrophages or DCs, in the long run, leads to persistently impaired functionality. TLR signals activate a wide range of negative feedback mechanisms; it is not known, however, which of these can lead to long-lasting tolerance for further stimulatory signals. In addition, it is not yet understood how the functionality of monocyte-derived DCs (MoDCs) is influenced in inflamed tissues by the continuous presence of stimulatory signals during their differentiation. Here we studied the role of a wide range of DC-inhibitory mechanisms in a simple and robust model of MoDC inactivation induced by early TLR signals during differentiation. We show that the activation-induced suppressor of cytokine signaling 1 (SOCS1), IL-10, STAT3, miR146a and CD150 (SLAM) molecules possessed short-term inhibitory effects on cytokine production but did not induce persistent DC inactivation. On the contrary, the LPS-induced IRAK-1 downregulation could alone lead to persistent MoDC inactivation. Studying cellular functions in line with the activation-induced negative feedback mechanisms, we show that early activation of developing MoDCs allowed only a transient cytokine production that was followed by the downregulation of effector functions and the preservation of a tissue-resident non-migratory phenotype.
Collapse
Affiliation(s)
- Tünde Fekete
- Department of Immunology, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Paracrine inhibition of GM-CSF signaling by human cytomegalovirus in monocytes differentiating to dendritic cells. Blood 2011; 118:6783-92. [PMID: 22031867 DOI: 10.1182/blood-2011-02-337956] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A primary HCMV infection or virus reactivation may cause severe disease in hosts with a deficient immune system. The virus can disturb both innate and adaptive immunity by targeting dendritic cell (DC) functions. Monocytes, the precursors of DCs in vivo (MoDCs), are the primary targets of HCMV; they can also harbor latent virus. The DCs generated from infected monocytes (CMV-MoDCs) have an altered phenotype and functional defects. We have shown that CMV-MoDCs do not secrete IL-12 in response to lipopolysaccharide stimulation, cannot ingest dead cells, induce T(H)1 differentiation, or the proliferation of naive allogeneic CD4(+) T cells. We found that the GM-CSF signaling in an entire population of CMV-MoDCs was impaired, although only half of the cells were productively infected, and that IL-6 secretion and suppressors of cytokine signaling 3 induction contributed to this bystander effect. We also showed that MoDCs derived ex vivo from monocytes of viremic patients had the same altered phenotype as CMV-MoDCs, including decreased STAT5 phosphorylation, indicating defective GM-CSF signaling. We have thus described a new mechanism of HCMV-induced immunosupression, indicated how infection may disturb both GM-CSF-dependent physiologic processes and proposed GM-CSF-based therapeutic approaches.
Collapse
|
24
|
Alaniz L, Rizzo M, Garcia MG, Piccioni F, Aquino JB, Malvicini M, Atorrasagasti C, Bayo J, Echeverria I, Sarobe P, Mazzolini G. Low molecular weight hyaluronan preconditioning of tumor-pulsed dendritic cells increases their migratory ability and induces immunity against murine colorectal carcinoma. Cancer Immunol Immunother 2011; 60:1383-95. [PMID: 21638126 PMCID: PMC11028773 DOI: 10.1007/s00262-011-1036-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 05/12/2011] [Indexed: 10/18/2022]
Abstract
We have recently shown that systemic administration of low molecular weight hyaluronan (LMW HA) significantly reduces colorectal carcinoma (CRC) growth in vivo. The elicited response is partially mediated by activated dendritic cells (DC). To potentiate the ability of DC loaded with whole tumor lysate (DC/TL) to induce immunity against CRC in mice, we aimed to study the effects of preconditioning DC with LMW HA for therapeutic vaccination. LMW HA improved maturation of ex vivo generated DC, increased IL-12, decreased IL-10 production, and enhanced a MLR activity in vitro. Although TNF-α showed a similar capacity to mature DC, preconditioning of DC/TL with LMW HA increased their ability to migrate in vitro toward CCL19 and CCL-21 in a CD44- and a TLR4-independent manner; this effect was superior to Poly(I:C), LPS, or TNF-α and partially associated with an increase in the expression of CCR7. Importantly, LMW HA dramatically enhanced the in vivo DC recruitment to tumor-regional lymph nodes. When these LMW HA-treated CRC tumor lysate-pulsed DC (DC/TL/LMW HA) were administered to tumor-bearing mice, a potent antitumor response was observed when compared to DC pulsed with tumor lysate alone and matured with TNF-α. Then, we showed that splenocytes isolated from animals treated with DC/TL/LMW HA presented a higher proliferative capacity, increased IFN-γ production, and secreted lower levels of the immunosuppressive IL-10. Besides, increased specific CTL response was observed in DC/TL/LMW HA-treated animals and induced long-term protection against tumor recurrence. Our data show that LMW HA is superior to other agents at inducing DC migration; therefore, LMW HA could be considered a new adjuvant candidate in the preparation of DC-based anticancer vaccines with potent immunostimulatory properties.
Collapse
Affiliation(s)
- Laura Alaniz
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Manglio Rizzo
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
| | - Mariana G. Garcia
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Flavia Piccioni
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
| | - Jorge B. Aquino
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Mariana Malvicini
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
| | - Itziar Echeverria
- CIMA, University of Navarra, Avenida Pio XII 55, 31008 Pamplona, Spain
| | - Pablo Sarobe
- CIMA, University of Navarra, Avenida Pio XII 55, 31008 Pamplona, Spain
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, School of Medicine, Austral University, Avenida Presidente Perón 1500 (B1629ODT) Derqui-Pilar, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| |
Collapse
|
25
|
Valencia J, Hernández-López C, Martínez VG, Hidalgo L, Zapata AG, Vicente Á, Varas A, Sacedón R. Wnt5a skews dendritic cell differentiation to an unconventional phenotype with tolerogenic features. THE JOURNAL OF IMMUNOLOGY 2011; 187:4129-39. [PMID: 21918189 DOI: 10.4049/jimmunol.1101243] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) are critical regulators of immune responses that integrate signals from the innate and adaptive immune system and orchestrate T cell responses toward either immunity or tolerance. Growing evidence points to the Wnt signaling pathway as a pivotal piece in the immune balance and focuses on DCs as a direct target for their immunoregulatory role. Our results show that the increase in Wnt5a signaling during the differentiation of human DCs from monocytes alters their phenotype and compromises their subsequent capacity to mature in response to TLR-dependent stimuli. These Wnt5a-DCs produce scant amounts of IL-12p70 and TNF-α but increased levels of IL-10. Consequently, these Wnt5a-DCs have a reduced capacity to induce Th1 responses that promote IL-10 secretion by CD4 T cells. Changes in the transcriptional profile of Wnt5a-DCs correlate with their unconventional phenotype caused presumably by increased IL-6/IL-10 signaling during the process of DC differentiation. The effect of Wnt5a is not a consequence of β-catenin accumulation but is dependent on noncanonical Ca(2+)/calmodulin-dependent protein kinase II/NF-κB signaling. Our results therefore suggest that under high levels of Wnt5a, typical of the inflammatory state and sepsis, monocytes could differentiate into unconventional DCs with tolerogenic features.
Collapse
Affiliation(s)
- Jaris Valencia
- Department of Cell Biology, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Fujimoto A, Akifusa S, Hirofuji T, Yamashita Y. Involvement of suppressor of cytokine signaling-1 in globular adiponectin-induced granulocyte colony-stimulating factor in RAW 264 cell. Mol Immunol 2011; 48:2052-8. [DOI: 10.1016/j.molimm.2011.06.440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 06/14/2011] [Accepted: 06/21/2011] [Indexed: 01/03/2023]
|
27
|
Puddu P, Latorre D, Carollo M, Catizone A, Ricci G, Valenti P, Gessani S. Bovine lactoferrin counteracts Toll-like receptor mediated activation signals in antigen presenting cells. PLoS One 2011; 6:e22504. [PMID: 21799877 PMCID: PMC3143167 DOI: 10.1371/journal.pone.0022504] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 06/27/2011] [Indexed: 01/17/2023] Open
Abstract
Lactoferrin (LF), a key element in mammalian immune system, plays pivotal roles in host defence against infection and excessive inflammation. Its protective effects range from direct antimicrobial activities against a large panel of microbes, including bacteria, viruses, fungi and parasites, to antinflammatory and anticancer activities. In this study, we show that monocyte-derived dendritic cells (MD-DCs) generated in the presence of bovine LF (bLF) fail to undergo activation by up-modulating CD83, co-stimulatory and major histocompatibility complex molecules, and cytokine/chemokine secretion. Moreover, these cells are weak activators of T cell proliferation and retain antigen uptake activity. Consistent with an impaired maturation, bLF-MD-DC primed T lymphocytes exhibit a functional unresponsiveness characterized by reduced expression of CD154 and impaired expression of IFN-γ and IL-2. The observed imunosuppressive effects correlate with an increased expression of molecules with negative regulatory functions (i.e. immunoglobulin-like transcript 3 and programmed death ligand 1), indoleamine 2,3-dioxygenase, and suppressor of cytokine signaling-3. Interestingly, bLF-MD-DCs produce IL-6 and exhibit constitutive signal transducer and activator of transcription 3 activation. Conversely, bLF exposure of already differentiated MD-DCs completely fails to induce IL-6, and partially inhibits Toll-like receptor (TLR) agonist-induced activation. Cell-specific differences in bLF internalization likely account for the distinct response elicited by bLF in monocytes versus immature DCs, providing a mechanistic base for its multiple effects. These results indicate that bLF exerts a potent anti-inflammatory activity by skewing monocyte differentiation into DCs with impaired capacity to undergo activation and to promote Th1 responses. Overall, these bLF-mediated effects may represent a strategy to block excessive DC activation upon TLR-induced inflammation, adding further evidence for a critical role of bLF in directing host immune function.
Collapse
Affiliation(s)
- Patrizia Puddu
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Daniela Latorre
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Carollo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Angela Catizone
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza, University of Rome, Rome, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Piera Valenti
- Department of Public Health Sciences and Infectious Diseases, Sapienza, University of Rome, Rome, Italy
| | - Sandra Gessani
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
- * E-mail:
| |
Collapse
|
28
|
Ray P, Arora M, Poe SL, Ray A. Lung myeloid-derived suppressor cells and regulation of inflammation. Immunol Res 2011; 50:153-8. [DOI: 10.1007/s12026-011-8230-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
29
|
O'Sullivan LA, Noor SM, Trengove MC, Lewis RS, Liongue C, Sprigg NS, Nicholson SE, Ward AC. Suppressor of cytokine signaling 1 regulates embryonic myelopoiesis independently of its effects on T cell development. THE JOURNAL OF IMMUNOLOGY 2011; 186:4751-61. [PMID: 21421851 DOI: 10.4049/jimmunol.1000343] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Suppressor of cytokine signaling 1 (SOCS1) has been shown to play important roles in the immune system. It acts as a key negative regulator of signaling via receptors for IFNs and other cytokines controlling T cell development, as well as Toll receptor signaling in macrophages and other immune cells. To gain further insight into SOCS1, we have identified and characterized the zebrafish socs1 gene, which exhibited sequence and functional conservation with its mammalian counterparts. Initially maternally derived, the socs1 gene showed early zygotic expression in mesodermal structures, including the posterior intermediate cell mass, a site of primitive hematopoiesis. At later time points, expression was seen in a broad anterior domain, liver, notochord, and intersegmental vesicles. Morpholino-mediated knockdown of socs1 resulted in perturbation of specific hematopoietic populations prior to the commencement of lymphopoiesis, ruling out T cell involvement. However, socs1 knockdown also lead to a reduction in the size of the developing thymus later in embryogenesis. Zebrafish SOCS1 was shown to be able to interact with both zebrafish Jak2a and Stat5.1 in vitro and in vivo. These studies demonstrate a conserved role for SOCS1 in T cell development and suggest a novel T cell-independent function in embryonic myelopoiesis mediated, at least in part, via its effects on receptors using the Jak2-Stat5 pathway.
Collapse
Affiliation(s)
- Lynda A O'Sullivan
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Wölfle SJ, Strebovsky J, Bartz H, Sähr A, Arnold C, Kaiser C, Dalpke AH, Heeg K. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur J Immunol 2011; 41:413-24. [PMID: 21268011 DOI: 10.1002/eji.201040979] [Citation(s) in RCA: 267] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 11/04/2010] [Accepted: 11/25/2010] [Indexed: 12/18/2022]
Abstract
During infection, TLR agonists are released and trigger mature as well as differentiating innate immune cells. Early encounter with TLR agonists (R848; LPS) blocks conventional differentiation of CD14(+) monocytes into immature dendritic cells (iDCs) resulting in a deviated phenotype. We and others characterized these APCs (TLR-APC) by a retained expression of CD14 and a lack of CD1a. Here, we show in addition, expression of programmed death ligand-1 (PD-L1). TLR-APCs failed to induce T-cell proliferation and furthermore were able to induce CD25(+) Foxp3(+) T regulatory cells (Tregs). Since PD-L1 is described as a key negative regulator and inducer of tolerance, we further analyzed its regulation. PD-L1 expression was regulated in a MAPK/cytokine/STAT-3-dependent manner: high levels of IL-6 and IL-10 that signal via STAT-3 were produced by TLR-APCs. Blocking of STAT-3 activation prevented PD-L1 expression. Moreover, chromatin immunoprecipitation revealed direct binding of STAT-3 to the PD-L1 promoter. Those findings indicate a pivotal role of STAT-3 in regulating PD-L1 expression. MAPKs were indirectly engaged, as blocking of p38 and p44/42 MAPKs decreased IL-6 and IL-10 thus reducing STAT-3 activation and subsequent PD-L1 expression. Hence, during DC differentiation TLR agonists induce a STAT-3-mediated expression of PD-L1 and favor the development of tolerogenic APCs.
Collapse
Affiliation(s)
- Sabine J Wölfle
- Department for Infectious Diseases, Medical Microbiology and Hygiene, University of Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Liontos LM, Dissanayake D, Ohashi PS, Weiss A, Dragone LL, McGlade CJ. The Src-Like Adaptor Protein Regulates GM-CSFR Signaling and Monocytic Dendritic Cell Maturation. THE JOURNAL OF IMMUNOLOGY 2011; 186:1923-33. [DOI: 10.4049/jimmunol.0903292] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
32
|
Downes JE, Marshall-Clarke S. Innate immune stimuli modulate bone marrow-derived dendritic cell production in vitro by toll-like receptor-dependent and -independent mechanisms. Immunology 2010; 131:513-24. [PMID: 20673241 PMCID: PMC2999802 DOI: 10.1111/j.1365-2567.2010.03324.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 05/27/2010] [Accepted: 06/03/2010] [Indexed: 12/27/2022] Open
Abstract
Haematopoiesis is crucial for immunity because it results in the production of leucocytes. Bacterial and viral infections alter leucocyte production by promoting granulopoiesis or lymphopoiesis. Recent studies suggest that changes in leucocyte production may be caused by the effects of inflammatory responses on the differentiation of haematopoietic progenitors in the bone marrow. We investigated the mechanisms through which infection regulates the formation of bone marrow-derived dendritic cells (BMDCs) in vitro. We mimicked infection by stimulating developing cells with molecules associated with bacteria and viruses and with inactivated influenza viruses. We showed that toll-like receptor (TLR) ligands act as modulators of haematopoiesis, and that signalling through different TLRs results in differing effects on the production of BMDCs. We demonstrated that ligands for TLR3 and influenza viruses reduce the production of BMDCs, resulting in increased neutrophil numbers, and that ligands for TLR4 and TLR9 drive the production of plasmacytoid dendritic cells. Furthermore, there are distinct signalling mechanisms involved in these effects. Signalling pathways triggered by TLR4 and TLR9 involve MyD88 and are partially mediated by the cytokine tumour necrosis factor-α (TNF-α). Mechanisms activated by TLR3 were Tir-domain-containing adaptor-inducing interferon dependent. Haematopoietic modulation induced by inactivated influenza viruses was associated with the activation of an antiviral pathway mediated by type-1 interferons.
Collapse
Affiliation(s)
- Joan E Downes
- Department of Human Anatomy and Cell Biology, The University of Liverpool, Liverpool, UK
| | | |
Collapse
|
33
|
Strebovsky J, Walker P, Lang R, Dalpke AH. Suppressor of cytokine signaling 1 (SOCS1) limits
NFκB
signaling by decreasing p65 stability within the cell nucleus. FASEB J 2010; 25:863-74. [DOI: 10.1096/fj.10-170597] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Julia Strebovsky
- Department of Infectious Diseases–Medical Microbiology and Hygiene University of Heidelberg Heidelberg Germany
| | - Patrick Walker
- Department of Infectious Diseases–Medical Microbiology and Hygiene University of Heidelberg Heidelberg Germany
| | - Roland Lang
- Institute of Clinical Microbiology Immunology and Hygiene University Hospital Erlangen Erlangen Germany
| | - Alexander H. Dalpke
- Department of Infectious Diseases–Medical Microbiology and Hygiene University of Heidelberg Heidelberg Germany
| |
Collapse
|
34
|
Subramanya S, Armant M, Salkowitz JR, Nyakeriga AM, Haridas V, Hasan M, Bansal A, Goepfert PA, Wynn KK, Ladell K, Price DA, N M, Kan-Mitchell J, Shankar P. Enhanced induction of HIV-specific cytotoxic T lymphocytes by dendritic cell-targeted delivery of SOCS-1 siRNA. Mol Ther 2010; 18:2028-37. [PMID: 20648001 PMCID: PMC2990509 DOI: 10.1038/mt.2010.148] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 06/15/2010] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that play a critical role in the activation of T cells. RNA interference (RNAi)-mediated silencing of negative immunoregulatory molecules expressed by DCs may provide a strategy to enhance the potency of DC-based vaccines and immunotherapy. Ablation of suppressor of cytokine signaling-1 (SOCS-1) in antigen-presenting cells has been shown to enhance cellular immune response in mice. Here, we used a previously reported DC-targeting approach to deliver small interfering RNA (siRNA) against SOCS-1 to human myeloid-derived DCs (MDDCs). SOCS1-silencing in MDDCs resulted in enhanced cytokine responses to lipopolysaccharide (LPS) and a strong mixed-lymphocyte reaction. Moreover, only DCs treated with SOCS-1 siRNA, and not controls, elicited strong primary in vitro responses to well-characterized HLA-A*0201-restricted Melan-A/MART-1 and human immunodeficiency virus (HIV) Gag epitopes in naive CD8(+) T cells from healthy donors. Finally, stimulation of CD8(+) T cells from HIV-seropositive subjects with SOCS1-silenced DCs resulted in an augmented polyfunctional cytotoxic T-lymphocyte (CTL) response, suggesting that SOCS-1 silencing can restore functionally compromised T cells in HIV infection. Collectively, these results demonstrate the feasibility of DC3-9dR-mediated manipulation of DC function to enhance DC immunogenicity for potential vaccine or immunotherapeutic applications.
Collapse
Affiliation(s)
- Sandesh Subramanya
- Immune Disease Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Targeting Toll-like receptor 7/8 enhances uptake of apoptotic leukemic cells by monocyte-derived dendritic cells but interferes with subsequent cytokine-induced maturation. Cancer Immunol Immunother 2010; 60:37-47. [PMID: 20859626 PMCID: PMC3029806 DOI: 10.1007/s00262-010-0917-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 08/31/2010] [Indexed: 12/13/2022]
Abstract
Therapeutic vaccination with dendritic cells (DC) is an emerging investigational therapy for eradication of minimal residual disease in acute myeloid leukemia. Various strategies are being explored in manufacturing DC vaccines ex vivo, e.g., monocyte-derived DC (MoDC) loaded with leukemia-associated antigens (LAA). However, the optimal source of LAA and the choice of DC-activating stimuli are still not well defined. Here, loading with leukemic cell preparations (harboring both unknown and known LAA) was explored in combination with a DC maturation-inducing cytokine cocktail (CC; IL-1β, IL-6, TNF-α, and PGE2) and Toll-like receptor ligands (TLR-L) to optimize uptake. Since heat shock induced apoptotic blasts were more efficiently taken up than lysates, we focused on uptake of apoptotic leukemic cells. Uptake of apoptotic blast was further enhanced by the TLR7/8-L R848 (20–30%); in contrast, CC-induced maturation inhibited uptake. CC, and to a lesser extent R848, enhanced the ability of MoDC to migrate and stimulate T cells. Furthermore, class II-associated invariant chain peptide expression was down-modulated after R848- or CC-induced maturation, indicating enhanced processing and presentation of antigenic peptides. To improve both uptake and maturation, leukemic cells and MoDC were co-incubated with R848 for 24 h followed by addition of CC. However, this approach interfered with CC-mediated MoDC maturation as indicated by diminished migratory and T cell stimulatory capacity, and the absence of IL-12 production. Taken together, our data demonstrate that even though R848 improved uptake of apoptotic leukemic cells, the sequential use of R848 and CC is counter-indicated due to its adverse effects on MoDC maturation.
Collapse
|
36
|
Shibata S, Maeda S, Kondo N, Inoue A, Maeda S, Chimura N, Fukata T. Effect of recombinant canine interferon-γ on granulocyte-macrophage colony-stimulating factor, transforming growth factor-β and CC chemokine ligand 17 mRNA transcription in a canine keratinocyte cell line (CPEK). Vet Dermatol 2010; 22:24-30. [DOI: 10.1111/j.1365-3164.2010.00897.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
37
|
Rydström A, Wick MJ. Salmonella inhibits monocyte differentiation into CD11c hi MHC-II hi cells in a MyD88-dependent fashion. J Leukoc Biol 2010; 87:823-32. [PMID: 20124491 DOI: 10.1189/jlb.0909615] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Monocytes and DCs originate from a shared precursor in the bone marrow, and steady-state DCs in lymphoid organs develop directly from the precursor rather than via a monocyte intermediate. However, monocytes can differentiate into DCs in tissues such as the lung and gut mucosa and into macrophages in most tissues. As Ly6C hi monocytes accumulate in lymphoid organs during oral Salmonella infection, we investigated their ability to develop into potential DCs, identified as CD11c hi MHC-II hi cells, in infected hosts. Ly6C hi monocytes, isolated from the blood of Salmonella-infected mice, developed into CD11c hi MHC-II hi cells after culture with GM-CSF or Flt3L. In contrast, the same monocytes cultured in the presence of GM-CSF and heat-killed Salmonella did not differentiate into CD11c hi MHC-II hi cells. The bacteria-induced differentiation block was dependent on TLRs, as monocytes from MyD88-/- mice converted into CD11c hi MHC-II hi cells even in the presence of bacteria. We hypothesized that Salmonella-activated wild-type monocytes secreted mediators that inhibited differentiation of MyD88-/--derived monocytes. However, IL-6, IL-10, TNF-alpha, or IL-12p70 did not account for the inhibition. Finally, monocyte-derived CD11c hi MHC-II hi cells pulsed with OVA peptide or protein did not induce proliferation of antigen-specific CD4+ T cells but rather, suppressed the ability of DCs to activate CD4+ T cells. Overall, the data show that Ly6C hi monocytes from Salmonella-infected mice develop into CD11c hi MHC-II hi cells with poor antigen-presentation capacity when cultured ex vivo, and that monocyte exposure to Salmonella inhibits their differentiation into CD11c hi MHC-II hi cells in a MyD88-dependent fashion.
Collapse
Affiliation(s)
- Anna Rydström
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
38
|
Oshansky CM, Krunkosky TM, Barber J, Jones LP, Tripp RA. Respiratory Syncytial Virus Proteins Modulate Suppressors of Cytokine Signaling 1 and 3 and the Type I Interferon Response to Infection by a Toll-Like Receptor Pathway. Viral Immunol 2009; 22:147-61. [DOI: 10.1089/vim.2008.0098] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Christine M. Oshansky
- Department of Infectious Diseases, College of Veterinary Medicine, Center for Disease Intervention, University of Georgia, Athens, Georgia
| | - Thomas M. Krunkosky
- Department of Anatomy and Radiology, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - Jamie Barber
- Department of Infectious Diseases, College of Veterinary Medicine, Center for Disease Intervention, University of Georgia, Athens, Georgia
| | - Les P. Jones
- Department of Infectious Diseases, College of Veterinary Medicine, Center for Disease Intervention, University of Georgia, Athens, Georgia
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, Center for Disease Intervention, University of Georgia, Athens, Georgia
| |
Collapse
|
39
|
Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, Grizzle WE, Mobley J, Zhang HG. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 2009; 124:2621-33. [PMID: 19235923 DOI: 10.1002/ijc.24249] [Citation(s) in RCA: 458] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) promote tumor progression. The mechanisms of MDSC development during tumor growth remain unknown. Tumor exosomes (T-exosomes) have been implicated to play a role in immune regulation, however the role of exosomes in the induction of MDSCs is unclear. Our previous work demonstrated that exosomes isolated from tumor cells are taken up by bone marrow myeloid cells. Here, we extend those findings showing that exosomes isolated from T-exosomes switch the differentiation pathway of these myeloid cells to the MDSC pathway (CD11b(+)Gr-1(+)). The resulting cells exhibit MDSC phenotypic and functional characteristics including promotion of tumor growth. Furthermore, we demonstrated that in vivo MDSC mediated promotion of tumor progression is dependent on T-exosome prostaglandin E2 (PGE2) and TGF-beta molecules. T-exosomes can induce the accumulation of MDSCs expressing Cox2, IL-6, VEGF, and arginase-1. Antibodies against exosomal PGE2 and TGF-beta block the activity of these exosomes on MDSC induction and therefore attenuate MDSC-mediated tumor-promoting ability. Exosomal PGE2 and TGF-beta are enriched in T-exosomes when compared with exosomes isolated from the supernatants of cultured tumor cells (C-exosomes). The tumor microenvironment has an effect on the potency of T-exosome mediated induction of MDSCs by regulating the sorting and the amount of exosomal PGE2 and TGF-beta available. Together, these findings lend themselves to developing specific targetable therapeutic strategies to reduce or eliminate MDSC-induced immunosuppression and hence enhance host antitumor immunotherapy efficacy.
Collapse
Affiliation(s)
- Xiaoyu Xiang
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Davies JM, Sheil B, Shanahan F. Bacterial signalling overrides cytokine signalling and modifies dendritic cell differentiation. Immunology 2009; 128:e805-15. [PMID: 19740342 DOI: 10.1111/j.1365-2567.2009.03086.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Heterogeneity of dendritic cells (DC) is evident in the gut-associated lymphoid tissue and determined, in part, by incompletely understood local environmental factors. Bacterial signalling is likely to be a dominant influence on precursor cells when recruited to the mucosa. We assessed the influence of commensal bacteria on DC differentiation and function. Murine bone marrow progenitors were exposed to Lactobacillus salivarius, Bifidobacterium breve or Bifidobacterium infantis. Differences in cell surface phenotype and function were assessed. Myeloid differentiation factor 88(-/-) (MyD88) cells were used to determine the influence of Toll-like receptor signalling. While bacterial strains varied in impact, there was a consistent dose-dependent inhibition of DC differentiation with a shift toward a Gr-1(+) CD11b(+) monocyte-like phenotype. A single bacterium on a per cell basis (1 : 1) was sufficient to alter cell phenotype. The effect was only evident in early precursors. Enhanced interleukin-10 production correlated with increased Forkhead box P3 expression and reduced T-cell proliferation. The bacterial effect on DC differentiation was found to be MyD88-dependent. Signalling by enteric commensals through pattern recognition receptors on precursor cells alters DC differentiation and results in cells that are phenotypically monocyte-like and functionally suppressive. This may account for some of the features of mucosal immune tolerance to the microbiota.
Collapse
Affiliation(s)
- Julie M Davies
- Alimentary Pharmabiotic Centre, University College Cork, National University of Ireland, Cork, Ireland
| | | | | |
Collapse
|
41
|
Ciaramella A, Sanarico N, Bizzoni F, Moro ML, Salani F, Scapigliati G, Spalletta G, Caltagirone C, Bossù P. Amyloid β peptide promotes differentiation of pro-inflammatory human myeloid dendritic cells. Neurobiol Aging 2009; 30:210-21. [PMID: 17658667 DOI: 10.1016/j.neurobiolaging.2007.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 06/11/2007] [Accepted: 06/12/2007] [Indexed: 12/25/2022]
Abstract
A key event of Alzheimer's disease (AD) pathogenesis is the production of amyloid beta peptides (A beta), which are hypothesized to lead to neurodegeneration by still unclear mechanisms, including a chronic inflammatory response characterized by innate immune cell activation and pro-inflammatory molecule release. Since dendritic cells (DCs) are central players of innate immune response and brain dendritic-like cells may have a crucial role in AD pathogenesis, this study investigates the effects of A beta on human DC functions. Myeloid DCs differentiated in the presence of A beta 42 showed an increase in survival and soluble antigen uptake, a reduction in HLA molecule expression and in IL-10 and IL-12 production. Accordingly, A beta 42-treated DCs were impaired in inducing T cell proliferation and IL-2 production. On the other hand, A beta 42 treatment provided DCs with the ability to release higher levels of IL-1 beta, IL-6 and IL-18, than control DCs. These results demonstrate that A beta 42 can modulate the immune system by inducing pro-inflammatory DC differentiation, thus gaining new insights into AD pathogenesis and immune-based therapeutic intervention.
Collapse
Affiliation(s)
- Antonio Ciaramella
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pène F, Zuber B, Courtine E, Rousseau C, Ouaaz F, Toubiana J, Tazi A, Mira JP, Chiche JD. Dendritic cells modulate lung response to Pseudomonas aeruginosa in a murine model of sepsis-induced immune dysfunction. THE JOURNAL OF IMMUNOLOGY 2009; 181:8513-20. [PMID: 19050269 DOI: 10.4049/jimmunol.181.12.8513] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Host infection by pathogens triggers an innate immune response leading to a systemic inflammatory response, often followed by an immune dysfunction which can favor the emergence of secondary infections. Dendritic cells (DCs) link innate and adaptive immunity and may be centrally involved in the regulation of sepsis-induced immune dysfunction. We assessed the contribution of DCs to lung defense in a murine model of sublethal polymicrobial sepsis (cecal ligature and puncture, CLP). In this model, bone marrow-derived DCs (BMDCs) retained an immature phenotype, associated with decreased capacity of IL-12p70 release and impaired priming of T cell lymphocytes. Eight days after CLP surgery, we induced a secondary pulmonary infection through intratracheal instillation of 5 x 10(6) CFUs of Pseudomonas aeruginosa. Whereas all sham-operated mice survived, 80% of post-CLP mice died after secondary pneumonia. Post-CLP mice exhibited marked lung damage with early recruitment of neutrophils, cytokine imbalance with decreased IL-12p70 production, and increased IL-10 release, but no defective bacterial lung clearance, while systemic bacterial dissemination was almost constant. Concomitant intrapulmonary administration of exogenous BMDCs into post-CLP mice challenged with P. aeruginosa dramatically improved survival. BMDCs did not improve bacterial lung clearance, but delayed neutrophil recruitment, strongly attenuated the early peak of TNF-alpha and restored an adequate Il-12p70/IL-10 balance in post-CLP mice. Thus, adoptive transfer of BMDCs reversed sepsis-induced immune dysfunction in a relevant model of secondary P. aeruginosa pneumonia. Unexpectedly, the mechanism of action of BMDCs did not involve enhanced antibacterial activity, but occurred by dampening the pulmonary inflammatory response.
Collapse
Affiliation(s)
- Frédéric Pène
- Department of Cellular Biology, Centre National de la Recherche Scientifique, Cochin Institute, University Paris-Descartes, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Seubert A, Monaci E, Pizza M, O'Hagan DT, Wack A. The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:5402-12. [PMID: 18390722 DOI: 10.4049/jimmunol.180.8.5402] [Citation(s) in RCA: 311] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aluminum hydroxide (alum) and the oil-in-water emulsion MF59 are widely used, safe and effective adjuvants, yet their mechanism of action is poorly understood. We assessed the effects of alum and MF59 on human immune cells and found that both induce secretion of chemokines, such as CCL2 (MCP-1), CCL3 (MIP-1alpha), CCL4 (MIP-1beta), and CXCL8 (IL-8), all involved in cell recruitment from blood into peripheral tissue. Alum appears to act mainly on macrophages and monocytes, whereas MF59 additionally targets granulocytes. Accordingly, monocytes and granulocytes migrate toward MF59-conditioned culture supernatants. In monocytes, both adjuvants lead to increased endocytosis, enhanced surface expression of MHC class II and CD86, and down-regulation of the monocyte marker CD14, which are all phenotypic changes consistent with a differentiation toward dendritic cells (DCs). When monocyte differentiation into DCs is induced by addition of cytokines, these adjuvants enhanced the acquisition of a mature DC phenotype and lead to an earlier and higher expression of MHC class II and CD86. In addition, MF59 induces further up-regulation of the maturation marker CD83 and the lymph node-homing receptor CCR7 on differentiating monocytes. Alum induces a similar but not identical pattern that clearly differs from the response to LPS. This model suggests a common adjuvant mechanism that is distinct from that mediated by danger signals. We conclude that during vaccination, adjuvants such as MF59 may increase recruitment of immune cells into the injection site, accelerate and enhance monocyte differentiation into DCs, augment Ag uptake, and facilitate migration of DCs into tissue-draining lymph nodes to prime adaptive immune responses.
Collapse
|
44
|
Katti MK, Dai G, Armitige LY, Marrero CR, Daniel S, Singh CR, Lindsey DR, Dhandayuthapani S, Hunter RL, Jagannath C. The Delta fbpA mutant derived from Mycobacterium tuberculosis H37Rv has an enhanced susceptibility to intracellular antimicrobial oxidative mechanisms, undergoes limited phagosome maturation and activates macrophages and dendritic cells. Cell Microbiol 2008; 10:1286-303. [PMID: 18248626 PMCID: PMC3668688 DOI: 10.1111/j.1462-5822.2008.01126.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mycobacterium tuberculosis H37Rv (Mtb) excludes phagocyte oxidase (phox) and inducible nitric oxide synthase (iNOS) while preventing lysosomal fusion in macrophages (MPhis). The antigen 85A deficient (Delta fbpA) mutant of Mtb was vaccinogenic in mice and the mechanisms of attenuation were compared with MPhis infected with H37Rv and BCG. Delta fbpA contained reduced amounts of trehalose 6, 6, dimycolate and induced minimal levels of SOCS-1 in MPhis. Blockade of oxidants enhanced the growth of Delta fbpA in MPhis that correlated with increased colocalization with phox and iNOS. Green fluorescent protein-expressing strains within MPhis or purified phagosomes were analysed for endosomal traffick with immunofluorescence and Western blot. Delta fbpA phagosomes were enriched for rab5, rab11, LAMP-1 and Hck suggesting enhanced fusion with early, recycling and late endosomes in MPhis compared with BCG or H37Rv. Delta fbpA phagosomes were thus more mature than H37Rv or BCG although, they failed to acquire rab7 and CD63 preventing lysosomal fusion. Finally, Delta fbpA infected MPhis and dendritic cells (DCs) showed an enhanced MHC-II and CD1d expression and primed immune T cells to release more IFN-gamma compared with those infected with BCG and H37Rv. Delta fbpA was thus more immunogenic in MPhis and DCs because of an enhanced susceptibility to oxidants and increased maturation.
Collapse
Affiliation(s)
- Muralidhar K. Katti
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Guixiang Dai
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Lisa Y. Armitige
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Carlos Rivera Marrero
- Department of Pulmonary Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Sundarsingh Daniel
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Christopher R. Singh
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Devin R. Lindsey
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | | | - Robert L. Hunter
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| |
Collapse
|
45
|
Abstract
Monocytes have been classically considered essential elements in relation with innate immune responses against pathogens, and inflammatory processes caused by external aggressions, infection and autoimmune disease. However, although their potential to differentiate into dendritic cells (DCs) was discovered 14 years ago, their functional relevance with regard to adaptive immune responses has only been uncovered very recently. Studies performed over the last years have revealed that monocyte-derived DCs play an important role in innate and adaptive immunity, due to their microbicidal potential, capacity to stimulate CD4(+) and CD8(+) T-cell responses and ability to regulate Immunoglobulin production by B cells. In addition, monocyte-derived DCs not only constitute a subset of DCs formed at inflammatory foci, as previously thought, but also comprise different subsets of DCs located in antigen capture areas, such as the skin and the intestinal, respiratory and reproductive tracts.
Collapse
|
46
|
Lin Y, Xiong S, Zhang L, Zhang Y, Cai Y, Xu L, Chu Y. Big tumor regression induced by GM-CSF gene-modified 3LL tumor cells via facilitating DC maturation and deviation toward CD11c+CD8alpha+ subset. DNA Cell Biol 2008; 26:863-72. [PMID: 17760559 DOI: 10.1089/dna.2007.0632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a powerful immune-stimulating factor that helps to generate a systemic, strong, and long-lasting immune response. However, whether the transduction of GM-CSF to tumor cell results in tumor regression and optimizes local immune microenvironment remains to be investigated. In this study, using an experimental murine tumor model, we demonstrated that the in vivo growth of 3LL tumor cells modified with the GM-CSF gene (3LL-GM) was inhibited even when the tumor diameter was over 7 mm (big tumor), and mice inoculated with GM-CSF gene-modified 3LL cells survived over 90 days, whereas mice inoculated with control parental 3LL cells and 3LL cells transduced with control vector all succumbed to the tumor by day 17 after tumor inoculation. Further analysis showed that targeted expression of GM-CSF in 3LL tumor cells markedly enhanced the systemic antitumor effect, including specific lymphocytes proliferation, cytotoxicity against 3LL tumor, and increased production of IFN-gamma. GM-CSF gene-modified 3LL cells significantly protected the mice from the parental 3LL tumor challenge. More importantly, the percentage of dendritic cells (DCs) in tumor site was greatly increased and the DCs differentiated into CD11c(+)CD8alpha(+) cells, which were reported to be able to benefit the induction of CD8(+) cytotoxic T lymphocytes (CTLs) that contribute to tumor regression. Our research indicated that GM-CSF could optimize the immune microenvironment in the tumor site, which provides a potent approach for immunotherapy of tumors.
Collapse
Affiliation(s)
- Yi Lin
- Department of Immunology, Institute for Immunobiology, Shanghai Medical College of Fudan University, Shanghai, P. R. China
| | | | | | | | | | | | | |
Collapse
|
47
|
Knuefermann P, Schwederski M, Velten M, Krings P, Ehrentraut H, Rüdiger M, Boehm O, Fink K, Dreiner U, Grohé C, Hoeft A, Baumgarten G, Koch A, Zacharowski K, Meyer R. Bacterial DNA induces myocardial inflammation and reduces cardiomyocyte contractility: role of toll-like receptor 9. Cardiovasc Res 2008; 78:26-35. [PMID: 18194990 DOI: 10.1093/cvr/cvn011] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS Myocardial function is severely compromised during sepsis. Several underlying mechanisms have been proposed. The innate immune system, i.e. toll-like receptor (TLR) 2 and 4, significantly contributes to cardiac dysfunction. Little is known regarding TLR9 and its pathogenic ligand bacterial DNA in the myocardium. We therefore studied the role of TLR9 in myocardial inflammation and cardiac contractility. METHODS AND RESULTS Wild-type (WT, C57BL/6) and TLR9-deficient (TLR9-D) mice and isolated cardiomyocytes were challenged with synthetic bacterial DNA (CpG-ODN). Myocardial contractility as well as markers of inflammation/signalling were determined. Isolated cardiomyocytes incorporated fluorescence-marked CpG-ODN. In WT mice, CpG-ODN caused a robust response in hearts demonstrated by increased levels of tumour necrosis factor (TNF-alpha), interleukin (IL)-1beta, IL-6, inducible nitric oxide synthase (iNOS), and nuclear factor kappaB activity. This inflammatory response was absent in TLR9-D mice. Under similar conditions, contractility measurements of isolated ventricular cardiomyocytes demonstrated a TLR9-dependent loss of sarcomeric shortening after CpG-ODN exposure. This observation was iNOS dependent as the application of a specific iNOS inhibitor reversed sarcomeric shortening to normal levels. CONCLUSION Our data suggest that bacterial DNA contributes to myocardial cytokine production and loss of cardiomyocyte contractility via TLR9.
Collapse
Affiliation(s)
- Pascal Knuefermann
- Department for Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Sigmund-Freud-Strasse 25, Bonn 53105, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dalpke A, Heeg K, Bartz H, Baetz A. Regulation of innate immunity by suppressor of cytokine signaling (SOCS) proteins. Immunobiology 2007; 213:225-35. [PMID: 18406369 DOI: 10.1016/j.imbio.2007.10.008] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 10/01/2007] [Accepted: 10/25/2007] [Indexed: 02/06/2023]
Abstract
Innate immunity represents the first line of defense against invading pathogens. Toll-like receptors (TLRs) are important for activation of innate immunity. Moreover, cytokines mediate communication of cells and are necessary to mount an appropriately regulated immune response. However, activation of innate immunity has to be tightly controlled to avoid overshooting immune reactions. Suppressor of cytokine signaling (SOCS) proteins have been identified as inducible feedback inhibitors of cytokine receptors and have been shown to be of crucial importance for the limitation of inflammatory responses. In this review, we describe the role of SOCS proteins in macrophages and dendritic cells (DCs). Based on our own findings, we show that SOCS proteins are directly induced by stimulation of TLRs. However, SOCS proteins do not interfere with direct TLR signaling, but avoid overshooting activation by regulating paracrine IFN-beta signaling. In addition, SOCS proteins in macrophages and DCs regulate the sensitivity towards IFN-gamma and GM-CSF, thereby modulating anti-microbial activity of macrophages and differentiation of DCs. We discuss that SOCS induction can also be used by microbes to evade immune defense, and this is exemplified by the parasite Toxoplasma gondii which induces SOCS1 to inhibit IFN-gamma-mediated macrophage activation. Taken together, the findings indicate that SOCS proteins play an important role in the balanced activation of innate immunity during infectious encounter.
Collapse
Affiliation(s)
- Alexander Dalpke
- Department of Hygiene and Medical Microbiology, Institute of Hygiene, University of Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | | | | | | |
Collapse
|
49
|
Yu S, Liu C, Su K, Wang J, Liu Y, Zhang L, Li C, Cong Y, Kimberly R, Grizzle WE, Falkson C, Zhang HG. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:6867-75. [PMID: 17513735 DOI: 10.4049/jimmunol.178.11.6867] [Citation(s) in RCA: 349] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The production of exosomes by tumor cells has been implicated in tumor-associated immune suppression. In this study, we show that, in mice, exosomes produced by TS/A murine mammary tumor cells target CD11b(+) myeloid precursors in the bone marrow (BM) in vivo, and that this is associated with an accumulation of myeloid precursors in the spleen. Moreover, we demonstrate that TS/A exosomes block the differentiation of murine myeloid precursor cells into dendritic cells (DC) in vitro. Addition of tumor exosomes at day 0 led to a significant block of differentiation into DC, whereas addition at later time points was less effective. Similarly, exosomes produced by human breast tumor cells inhibited the differentiation of human monocytes in vitro. The levels of IL-6 and phosphorylated Stat3 were elevated 12 h after the tumor exosome stimulation of murine myeloid precursors, and tumor exosomes were less effective in inhibiting differentiation of BM cells isolated from IL-6 knockout mice. Addition of a rIL-6 to the IL-6 knockout BM cell culture restored the tumor exosome-mediated inhibition of DC differentiation. These data suggest that tumor exosome-mediated induction of IL-6 plays a role in blocking BM DC differentiation.
Collapse
MESH Headings
- Adult
- Animals
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Cell Differentiation/immunology
- Cell Line, Tumor
- Cells, Cultured
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Exocytosis/immunology
- Female
- Growth Inhibitors/immunology
- Growth Inhibitors/metabolism
- Humans
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Myeloid Progenitor Cells/cytology
- Myeloid Progenitor Cells/immunology
- Secretory Vesicles/immunology
- Secretory Vesicles/metabolism
- Secretory Vesicles/pathology
Collapse
Affiliation(s)
- Shaohua Yu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Suppressor of cytokine signalling (SOCS) proteins are inhibitors of cytokine signalling pathways. Studies have shown that SOCS proteins are key physiological regulators of both innate and adaptive immunity. These molecules positively and negatively regulate macrophage and dendritic-cell activation and are essential for T-cell development and differentiation. Evidence is also emerging of the involvement of SOCS proteins in diseases of the immune system. In this Review we bring together data from recent studies on SOCS proteins and their role in immunity, and propose a cohesive model of how cytokine signalling regulates immune-cell function.
Collapse
Affiliation(s)
- Akihiko Yoshimura
- Division of Molecular and Cellular Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | | | | |
Collapse
|