Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Neuroinflammation ; 20(1): 307, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38124095

ABSTRACT

BACKGROUND: Reinstating inflammation resolution represents an innovative concept to regain inflammation control in diseases marked by chronic inflammation. While most therapeutics target inflammatory molecules and inflammatory effector cells and mediators, targeting macrophages to initiate inflammation resolution to control neuroinflammation has not yet been attempted. Resolution-phase macrophages are critical in the resolution process to regain tissue homeostasis, and are programmed through the presence and elimination of apoptotic leukocytes. Hence, inducing resolution-phase macrophages might represent an innovative therapeutic approach to control and terminate dysregulated neuroinflammation. METHODS: Here, we investigated if the factors released by in vitro induced resolution-phase macrophages (their secretome) are able to therapeutically reprogram macrophages to control neuroinflammation in the model of experimental autoimmune encephalomyelitis (EAE). RESULTS: We found that injection of the pro-resolutive secretome reduced demyelination and decreased inflammatory cell infiltration in the CNS, notably through the in vivo reprogramming of macrophages at the epigenetic level. Adoptive transfer experiments with in vivo or in vitro reprogrammed macrophages using such pro-resolutive secretome confirmed the stability and transferability of this acquired therapeutic activity. CONCLUSIONS: Overall, our data confirm the therapeutic activity of a pro-resolution secretome in the treatment of ongoing CNS inflammation, via the epigenetic reprogramming of macrophages and open with that a new therapeutic avenue for diseases marked by neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Animals , Neuroinflammatory Diseases , Macrophages , Inflammation , Leukocytes
2.
Front Immunol ; 12: 754475, 2021.
Article in English | MEDLINE | ID: mdl-35003066

ABSTRACT

Nonresolving inflammation is a critical driver of several chronic inflammatory diseases, including inflammatory bowel diseases (IBD). This unresolved inflammation may result from the persistence of an initiating stimulus or from the alteration of the resolution phase of inflammation. Elimination of apoptotic cells by macrophages (a process called efferocytosis) is a critical step in the resolution phase of inflammation. Efferocytosis participates in macrophage reprogramming and favors the release of numerous pro-resolving factors. These pro-resolving factors exert therapeutic effects in experimental autoimmune arthritis. Here, we propose to evaluate the efficacy of pro-resolving factors produced by macrophages after efferocytosis, a secretome called SuperMApo, in two IBD models, namely dextran sodium sulfate (DSS)-induced and T cell transfer-induced colitis. Reintroducing these pro-resolving factors was sufficient to decrease clinical, endoscopic and histological colitis scores in ongoing naive T cell-transfer-induced colitis and in DSS-induced colitis. Mouse primary fibroblasts isolated from the colon demonstrated enhanced healing properties in the presence of SuperMApo, as attested by their increased migratory, proliferative and contractive properties. This was confirmed by the use of human fibroblasts isolated from patients with IBD. Exposure of an intestinal epithelial cell (IEC) line to these pro-resolving factors increased their proliferative properties and IEC acquired the capacity to capture apoptotic cells. The improvement of wound healing properties induced by SuperMApo was confirmed in vivo in a biopsy forceps-wound colonic mucosa model. Further in vivo analysis in naive T cell transfer-induced colitis model demonstrated an improvement of intestinal barrier permeability after administration of SuperMApo, an intestinal cell proliferation and an increase of α-SMA expression by fibroblasts, as well as a reduction of the transcript coding for fibronectin (Fn1). Finally, we identified TGF-ß, IGF-I and VEGF among SuperMApo as necessary to favor mucosal healing and confirmed their role both in vitro (using neutralizing antibodies) and in vivo by depleting these factors from efferocytic macrophage secretome using antibody-coated microbeads. These growth factors only explained some of the beneficial effects induced by factors released by efferocytic macrophages. Overall, the administration of pro-resolving factors released by efferocytic macrophages limits intestinal inflammation and enhance tissue repair, which represents an innovative treatment of IBD.


Subject(s)
Biological Factors/physiology , Cytophagocytosis/physiology , Fibroblasts/physiology , Inflammatory Bowel Diseases/immunology , Macrophages/physiology , Wound Healing/physiology , Actins/biosynthesis , Actins/genetics , Animals , Biological Factors/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Division/drug effects , Cell Line , Colitis/chemically induced , Colitis/etiology , Colitis/immunology , DNA-Binding Proteins/deficiency , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Fibronectins/biosynthesis , Fibronectins/genetics , Humans , Inflammatory Bowel Diseases/physiopathology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/cytology , Intestinal Mucosa/injuries , Lymphocyte Transfusion/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms
3.
Front Immunol ; 9: 2586, 2018.
Article in English | MEDLINE | ID: mdl-30542342

ABSTRACT

Unresolved inflammation is a common feature in the pathogenesis of chronic inflammatory/autoimmune diseases. The factors produced by macrophages eliminating apoptotic cells during resolution are crucial to terminate inflammation, and for subsequent tissue healing. We demonstrated here that the factors produced by macrophages eliminating apoptotic cells were sufficient to reboot the resolution of inflammation in vivo, and thus definitively terminated ongoing chronic inflammation. These factors were called SuperMApo and revealed pro-resolutive properties and accelerated acute inflammation resolution, as attested by both increased phagocytic capacities of macrophages and enhanced thioglycollate-induced peritonitis resolution. Activated antigen-presenting cells exposed to SuperMApo accelerated their return to homeostasis and demonstrated pro-regulatory T cell properties. In mice with ongoing collagen-induced arthritis, SuperMApo injection resolved and definitively terminated chronic inflammation. The same pro-resolving properties were observed in human settings in addition to xenogeneic colitis and graft-vs.-host disease modulation, highlighting SuperMApo as a new therapeutic opportunity to circumvent inflammatory diseases.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Macrophages, Peritoneal/immunology , Animals , Antigen-Presenting Cells/immunology , Colitis/immunology , Female , Homeostasis/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Peritonitis/immunology , Phagocytosis/immunology , T-Lymphocytes, Regulatory/immunology
4.
Arthritis Res Ther ; 18(1): 184, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27516061

ABSTRACT

BACKGROUND: Apoptotic cell-based therapies have been proposed to treat chronic inflammatory diseases. The aim of this study was to investigate the effect of intravenous (i.v.) apoptotic cell infusion in ongoing collagen-induced arthritis (CIA) and the interaction of this therapy with other treatments used in rheumatoid arthritis (RA), including methotrexate (MTX) or anti-TNF therapy. METHODS: The effects of i.v. apoptotic cell infusion were evaluated in a CIA mouse model in DBA/1 mice immunized with bovine type II collagen. The number and functions of antigen-presenting cells (APC), regulatory CD4(+) T cells (Treg), and circulating anti-collagen auto-antibodies were analyzed in CIA mice. RESULTS: Treatment of arthritic mice with i.v. apoptotic cell infusion significantly reduced the arthritis clinical score. This therapeutic approach modified T cell responses against the collagen auto-antigen with selective induction of collagen-specific Treg. In addition, we observed that APC from apoptotic-cell-treated animals were resistant to toll-like receptor ligand activation and favored ex vivo Treg induction, indicating APC reprogramming. Apoptotic cell injection-induced arthritis modulation was dependent on transforming growth factor (TGF)-ß, as neutralizing anti-TGF-ß antibody prevented the effects of apoptotic cells. Methotrexate did not interfere, while anti-TNF therapy was synergic with apoptotic-cell-based therapy. CONCLUSION: Overall, our data demonstrate that apoptotic-cell-based therapy is efficient in treating ongoing CIA, compatible with current RA treatments, and needs to be evaluated in humans in the treatment of RA.


Subject(s)
Apoptosis/immunology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cell- and Tissue-Based Therapy/methods , Thymocytes/transplantation , Animals , Antirheumatic Agents/pharmacology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Female , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Tumor Necrosis Factor-alpha/antagonists & inhibitors
5.
J Immunol ; 197(5): 1672-82, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27448589

ABSTRACT

Regulation of the inflammatory response involves the control of dendritic cell survival. To our knowledge, nothing is known about the survival of plasmacytoid dendritic cells (pDC) in such situation. pDC are specialized in type I IFN (IFN-I) secretion to control viral infections, and IFN-I also negatively regulate pDC survival during the course of viral infections. In this study, we asked about pDC behavior in the setting of virus-free inflammation. We report that pDC survival was profoundly reduced during different nonviral inflammatory situations in the mouse, through a mechanism independent of IFN-I and TLR signaling. Indeed, we demonstrated that during inflammation, CD8(+) T cells induced pDC apoptosis through the perforin pathway. The data suggest, therefore, that pDC have to be turned down during ongoing acute inflammation to not initiate autoimmunity. Manipulating CD8(+) T cell response may therefore represent a new therapeutic opportunity for the treatment of pDC-associated autoimmune diseases, such as lupus or psoriasis.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Dendritic Cells/pathology , Perforin/metabolism , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/metabolism , Cytokines/blood , Disease Models, Animal , Graft vs Host Disease/immunology , Inflammation/immunology , Interferon-alpha/immunology , Interferon-alpha/metabolism , Mice , Perforin/immunology , Toll-Like Receptor 7/metabolism
6.
Transfusion ; 55(7): 1721-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25807899

ABSTRACT

BACKGROUND: Red blood cell (RBC) alloimmunization is a major immunologic risk of transfusion. However, RBC storage facilitates white blood cell (WBC) apoptosis and apoptotic cells have immunomodulatory properties. We investigated the behavior of WBCs, and apoptosis in particular, in RBC units during storage and then studied the impact of WBC apoptosis on the modulation of posttransfusion alloimmunization in RBC products stored short term. STUDY DESIGN AND METHODS: We used a mouse model of alloimmunization to transfused HEL-ovalbumin-Duffy (HOD) surface antigen expressed specifically on RBCs. The presence of circulating anti-HOD immunoglobulin G detected by flow cytometry confirmed immunization to HOD+ RBCs. WBC apoptosis and factors released by apoptotic WBCs during storage were determined and in particular the role of transforming growth factor (TGF)-ß was assessed on RBC alloimmunization. RESULTS: In blood stored 72 hours, 30% of WBCs were apoptotic, and transfusion of short-term-stored blood resulted in lesser immunization than did fresh blood or stored leukoreduced (LR) RBCs. WBCs undergoing apoptosis released during short-term storage factors modulating RBC alloimmunization. Indeed apoptotic cell-released factors modulate alloimmunization whereas exogenous apoptotic cells directly transfused with LR RBCs did not. While microparticles released during RBC storage had no immunomodulatory role, TGF-ß found in the supernatant of stored blood demonstrated the capacity to favor Treg polarization of naïve CD4+CD25- T cells in vitro and limited RBC alloimmunization in vivo. Indeed, addition of recombinant TGF-ß to stored LR RBC transfusion strongly limited posttransfusion RBC alloimmunization. CONCLUSION: Our findings show that short-term storage of non-LR blood facilitates WBC apoptosis therefore releasing TGF-ß that modulates posttransfusion RBC alloimmunization.


Subject(s)
Apoptosis/immunology , Blood Preservation , Erythrocyte Transfusion , Erythrocytes/immunology , Immunomodulation , Leukocytes/immunology , Transforming Growth Factor beta/immunology , Animals , Erythrocytes/metabolism , Leukocytes/metabolism , Mice , Mice, Inbred BALB C , Transforming Growth Factor beta/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...