Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Allergy Clin Immunol ; 136(2): 351-9.e1, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25792465

ABSTRACT

BACKGROUND: Psoriasis is a systemic inflammatory disease in which IL-17 and IL-22 levels are markedly increased in the skin and blood. The prevalent concept, using skin cells that are isolated from psoriatic plaques and examined after cell expansion and in vitro stimulation, is that IL-17 and IL-22 production essentially results from T cells and the rare type 3 innate lymphoid cells. OBJECTIVE: We sought to examine the cellular source of IL-17A and IL-22 at the protein and transcriptional single-cell level immediately after ex vivo skin cell isolation from psoriatic plaques. METHODS: Skin biopsy specimens were collected from patients with psoriasis, as well as from patients with atopic dermatitis. Cell suspensions were prepared by combining mild enzymatic digestion and mechanical dissociation and analyzed for cytokine expression without prior in vitro culture and stimulation. Expression of IL-17 and IL-22 was quantified at the protein and mRNA single-cell level by using flow cytometry. RESULTS: IL-22 is predominantly expressed by CD3(-)c-Kit(+) cells relative to CD3(+) T cells in lesional skin of patients with psoriasis and patients with atopic dermatitis. Strikingly, we identified c-Kit(+)FcεRI(+) mast cells as major IL-22 producers. The proportion of mast cells that produce IL-22 ranges from 20% to 80% in patients with psoriasis or those with atopic dermatitis. Skin mast cells express IL-22 and IL-17 mRNA. Conversely, IL-17-producing T cells outnumber IL-17-producing mast cells, which also express IL-17 receptor. CONCLUSION: Human skin mast cells are previously unrecognized IL-22 producers. We further established that skin mast cells express IL-17. Thus mast cells might play an important role in the physiopathology of chronic inflammatory skin disorders.


Subject(s)
Dermatitis, Atopic/immunology , Interleukin-17/immunology , Interleukins/immunology , Mast Cells/immunology , Psoriasis/immunology , T-Lymphocytes/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Count , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Gene Expression , Humans , Interleukin-17/genetics , Interleukins/genetics , Mast Cells/pathology , Primary Cell Culture , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Psoriasis/genetics , Psoriasis/pathology , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Skin/immunology , Skin/pathology , T-Lymphocytes/pathology , Interleukin-22
2.
Blood ; 120(24): 4761-71, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23071273

ABSTRACT

Basophils are a rare population of granulocytes that have long been associated with IgE-mediated and Th2-associated allergic diseases. However, the role of basophils in Th17 and/or Th1 diseases has not been reported. In the present study, we report that basophils can be detected in the mucosa of Th17-associated lung and inflammatory bowel disease and accumulate in inflamed colons containing large quantities of IL-33. We also demonstrate that circulating basophils increased memory Th17 responses. Accordingly, IL-3- or IL-33-activated basophils amplified IL-17 release in effector memory T cells (T(EM)), central memory T cells (T(CM)), and CCR6(+) CD4 T cells. More specifically, basophils promoted the emergence of IL-17(+)IFN-γ(-) and IL-17(+)IFN-γ(+), but not IL-17(-)IFN-γ(+) CD4 T cells in T(EM) and T(CM). Mechanistic analysis revealed that the enhancing effect of IL-17 production by basophils in T(EM) involved the ERK1/2 signaling pathway, occurred in a contact-independent manner, and was partially mediated by histamine via H(2) and H(4) histamine receptors. The results of the present study reveal a previously unknown function for basophils in augmenting Th17 and Th17/Th1 cytokine expression in memory CD4 T cells. Because basophils accumulated in inflamed inflammatory bowel disease tissues, we propose that these cells are key players in chronic inflammatory disorders beyond Th2.


Subject(s)
Basophils/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Basophils/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Flow Cytometry , Gene Expression/drug effects , Gene Expression/immunology , Histamine/immunology , Histamine/metabolism , Humans , Immunologic Memory/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-3/immunology , Interleukin-3/pharmacology , Interleukin-33 , Interleukins/immunology , Interleukins/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Receptors, Histamine H2/genetics , Receptors, Histamine H2/immunology , Reverse Transcriptase Polymerase Chain Reaction , Th17 Cells/drug effects , Th17 Cells/metabolism
3.
J Immunol ; 188(9): 4249-55, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22461697

ABSTRACT

T cell memory is the hallmark of adaptive immunity. Central questions are to determine which cells among proliferating effector T cells will live beyond the crash of the immune response (IR) and develop into functional memory T cells. CD47, considered as a marker of self, is implicated in cell death, cell elimination, and in the inflammatory response. We report in this article that CD47 expression was transiently regulated on Ag-specific CD4 T cells, that is, from CD47(high) to CD47(low) to CD47(high), during the course of the in vivo IR. Specifically, CD47(high) status marked central memory CD4 T cell precursors at an early time point of the IR. By contrast, cytokine production was a functional attribute restricted to CD47(high), but not CD47(low), polyclonal effector CD4 T cells during recall responses in an experimental model of chronic airway inflammatory disease. Passive transfer of CD47(high), but not CD47(low), CD4 T cells in nonlymphopenic naive mice generated long-lived memory T cells capable of anamnestic responses. We conclude that CD47(high) status on CD4 T cells identifies functional long-lived memory T cell progenitors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD47 Antigen/immunology , Gene Expression Regulation/physiology , Immunity, Cellular/physiology , Immunologic Memory/physiology , Precursor Cells, T-Lymphoid/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD47 Antigen/biosynthesis , CD47 Antigen/genetics , Mice , Mice, Knockout , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism
4.
Brain Dev ; 44(10): 681-689, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36050140

ABSTRACT

INTRODUCTION: Cerebral palsy (CP) is the most prevalent motor disorder of childhood. It typically results from in utero or perinatal brain injury. Recently, it has been reported that autologous cord blood (ACB) infusion for children with CP improved gross motor function and brain connectivity, but unfortunately, it has never been tried in Japan. We conducted a pilot study of the infusing of ACB, which was delivered from private bank, in the children with CP to assess the safety and feasibility to the procedure as well as any effect in improving neurological function. METHODS: After demonstrating the induction of tissue regeneration in animal model studies conducted a single-arm pilot study of intravenous ACB infusion in 6 young Japanese children with CP (ages 1-6 years). Primary outcomes were safety assessed by vital signs, clinical symptoms, and blood and urinary examinations at baseline and 1 weeks, 1, 2 and 3 years after treatment. In addition, motor function evaluations, neurodevelopmental examinations, magnetic resonance imaging, and electroencephalography (EEG) were performed at the same time. RESULTS: Infusion was generally well-tolerated, although one patient experienced microhematuria 1 year after treatment and another one patient experienced febrile convulsion once 9 months after treatment. These events were transient, no relapse was seen during observation study. All patients improved a median of 6.8 points on the 1-year Gross Motor Functional Measure-66 (GMFM-66) scores, greater than predicted by age and severity. Furthermore, the 2-year and 3-year GMFM-66 scores were also greater than expected (median 6.2 points and 5.5 points, respectively). Overall scales and language-social scales of the developmental quotient (DQ) improved in 3 of 6 patients, who had greater changes in their GMFM-66 scores than the other cases after treatment. There were no significant correlations among the GMFM-66 scores, DQ, and infusion cell counts. CONCLUSION: ACB infusion was safe and feasible for clinical use in patients with CP. However, much more clinical study with larger numbers of patients and in-depth studies of treatment mechanism of CP are needed.


Subject(s)
Cerebral Palsy , Humans , Cerebral Palsy/therapy , Fetal Blood , Japan , Feasibility Studies , Pilot Projects , Motor Skills
5.
Int Immunol ; 22(4): 237-44, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20181653

ABSTRACT

The balance between effector CD4(+) T cells secreting IL-17 (T(h)17) and regulatory T cells (Treg) plays an important role in autoimmune disorders that include rheumatoid arthritis (RA) and Crohn's disease. Tumor necrosis factor (TNF)-alpha is a key pro-inflammatory cytokine that contributes to disease pathogenesis. We investigated the interplay between CD45RA(+) Treg and TNF-alpha in the regulation of human T(h)17 differentiation. We found that CD45RA(+) Treg promoted while TNF-alpha inhibited naive CD4(+) T-cell differentiation into IL-17 and CCL20 co-expressing T(h)17 cells without influencing their IL-22 release. Unexpectedly, CD45RA(+) Treg depletion abrogated TNF-alpha suppressive function. Finally, dendritic cell-derived TNF-alpha suppressed the development of IL-17(+)CCL20(+) expressing T(h)17 cells. In conclusion, CD45RA(+) Treg positively governs human T(h)17 development, which is impaired by TNF-alpha. We propose that TNF-alpha may represent a negative feedback mechanism to control IL-17/CCL20- but not IL-22-associated autoimmune pathologies.


Subject(s)
Autoimmune Diseases/immunology , Leukocyte Common Antigens/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/immunology , Cell Differentiation , Cells, Cultured , Chemokine CCL20/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Leukocyte Common Antigens/biosynthesis , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Interleukin-22
7.
Immunology ; 128(1 Suppl): e523-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740313

ABSTRACT

Enteric infections remain a major health problem causing millions of deaths in developing countries. The interplay among the host intestinal epithelium, the mucosa-associated immune system and microbiota performs an essential role in gut homeostasis and protection against infectious diseases. Dendritic cells (DCs) play a key role in orchestrating protective immunity and tolerance in the gut. The mechanisms by which DCs adapt their responses and discriminate between virulent microbes and trillions of innocuous bacteria remain ill-defined. Here we investigated the effect of cross-talk between commensal-related bacteria (CB) and Toll-like receptor (TLR) agonists on DC activation and the outcome of the in vitro T helper response. Human monocyte-derived DCs were exposed to eight different Gram-positive or Gram-negative CB strains prior to activation with five different TLR agonists. The key polarizing cytokines interleukin (IL)-12p70, IL-10, IL-1beta and IL-6 were quantified and the fate of naïve T-cell differentiation was evaluated. We identified a unique combination of Lactobacillus casei and TLR3 signals that acted in synergy to selectively increase IL-12p70 secretion. Exposure to poly(I:C) converted L. casei-treated DCs into potent promoters of T helper type 1 (Th1) responses. We propose that DCs can integrate harmless and dangerous non-self signals delivered by viral products, to mount robust Th1 responses. Thus, in vivo DC targeting with selective probiotics may improve strategies for the management of enteric diseases.


Subject(s)
Dendritic Cells/immunology , Interleukin-12/biosynthesis , Intestines/immunology , Lacticaseibacillus casei/immunology , Probiotics , Th1 Cells/immunology , Toll-Like Receptor 3/agonists , Dendritic Cells/drug effects , Dendritic Cells/microbiology , Flagellin/immunology , Humans , Interferon Inducers/pharmacology , Interleukin-10/biosynthesis , Interleukin-12/agonists , Interleukin-12/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Interleukin-6/biosynthesis , Intestines/microbiology , Lipopolysaccharides/immunology , Peptidoglycan/immunology , Poly I-C/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Tetradecanoylphorbol Acetate/pharmacology , Th1 Cells/drug effects , Th1 Cells/microbiology , Toll-Like Receptor 3/metabolism
8.
J Leukoc Biol ; 84(2): 468-76, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511576

ABSTRACT

Dendritic cells (DCs) orchestrate the immune response establishing immunity versus tolerance. These two opposite functions may be dictated by DC maturation status with maturity linked to immunogenicity. DCs directly interact with trillions of noninvasive intestinal bacteria in vivo, a process that contributes to gut homeostasis. We here evaluated the maturation program elicited in human DCs by direct exposure to commensal-related bacteria (CB) in the absence of inflammatory signals. We showed that eight gram(+) and gram(-) CB strains up-regulated costimulatory molecule expression in DCs and provoked a chemokine receptor switch similar to that activated by gram(+) pathogens. CB strains may be classified into three groups according to DC cytokine release: high IL-12 and low IL-10; low IL-12 and high IL-10; and low IL-12 and IL-10. All CB-treated DCs produced IL-1beta and IL-6 and almost no TGF-beta. Yet, CB instructed DCs to convert naive CD4+ T cells into hyporesponsive T cells that secreted low or no IFN-gamma, IL-10, and IL-17 and instead, displayed suppressor function. These data demonstrate that phenotypic DC maturation combined to an appropriate cytokine profile is insufficient to warrant Th1, IL-10-secreting T regulatory Type 1 (Tr1), or Th17 polarization. We propose that commensal flora and as such, probiotics manipulate DCs by a yet-unidentified pathway to enforce gut tolerance.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/microbiology , Lacticaseibacillus casei/immunology , T-Lymphocytes, Regulatory/immunology , Antibody Formation , Cell Culture Techniques , Flow Cytometry , Humans , Immunosuppression Therapy , Interleukin-10/metabolism , Interleukin-12/metabolism , Tissue Expansion
9.
PLoS One ; 14(9): e0221111, 2019.
Article in English | MEDLINE | ID: mdl-31483787

ABSTRACT

Regenerative medicine using umbilical cord blood (UCB) cells shows promise for the treatment of cerebral palsy. Although the efficacy of this therapy has been seen in the clinic, the mechanisms by which UCB cells interact and aid in the improvement of symptoms are not clear. We explored the chemokine expression profile in damaged brain tissue in the neonatal mouse ischemia-reperfusion (IR) brain injury model that was infused with human UCB (hUCB) cells. IR brain injury was induced in 9-day-old NOD/SCID mice. hUCB cells were administered 3 weeks post brain injury. Chemokine expression profiles in the brain extract were determined at various time points. Inflammatory chemokines such as CCL1, CCL17, and CXCL12 were transiently upregulated by 24 hours post brain injury. Upregulation of other chemokines, including CCL5, CCL9, and CXCL1 were prolonged up to 3 weeks post brain injury, but most chemokines dissipated over time. There were marked increases in levels of CCL2, CCL12, CCL20, and CX3CL1 in response to hUCB cell treatment, which might be related to the new recruitment and differentiation of neural stem cells, leading to the induction of tissue regeneration. We propose that the chemokine expression profile in the brain shifted from responding to tissue damage to inducing tissue regeneration. hUCB cell administration further enhanced the production of chemokines, and chemokine networks may play an active role in tissue regeneration in neonatal hypoxic-ischemic brain injury.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Chemokines/genetics , Cord Blood Stem Cell Transplantation , Fetal Blood/cytology , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Animals , Biomarkers , Brain Injuries/therapy , Chemokines/metabolism , Cord Blood Stem Cell Transplantation/methods , Cytokines/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Reperfusion Injury/therapy
10.
Stem Cell Res Ther ; 8(1): 26, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28173860

ABSTRACT

BACKGROUND: Neonatal hypoxia-ischemia induces massive brain damage during the perinatal period, resulting in long-term consequences to central nervous system structural and functional maturation. Although neural progenitor cells (NPCs) migrate through the parenchyma and home in to injury sites in the rodent brain, the molecular mechanisms are unknown. We examined the role of chemokines in mediating NPC migration after neonatal hypoxic-ischemic brain injury. METHODS: Nine-day-old mice were exposed to a 120-minute hypoxia following unilateral carotid occlusion. Chemokine levels were quantified in mouse brain extract. Migration and proliferation assays were performed using embryonic and infant mouse NPCs. RESULTS: The neonatal hypoxic-ischemic brain injury resulted in an ipsilateral lesion, which was extended to the cortical and striatal areas. NPCs migrated toward an injured area, where a marked increase of CC chemokines was detected. In vitro studies showed that incubation of NPCs with recombinant mouse CCL11 promoted migration and proliferation. These effects were partly inhibited by a CCR3 antagonist, SB297006. CONCLUSIONS: Our data implicate an important effect of CCL11 for mouse NPCs. The effective activation of NPCs may offer a promising strategy for neuroregeneration in neonatal hypoxic-ischemic brain injury.


Subject(s)
Brain Injuries/pathology , Brain/pathology , Chemokine CCL11/pharmacology , Hypoxia-Ischemia, Brain/pathology , Neural Stem Cells/drug effects , Animals , Animals, Newborn , Benzamides/pharmacology , Brain/metabolism , Brain Injuries/metabolism , Carotid Arteries/surgery , Cell Movement/drug effects , Cell Proliferation/drug effects , Cerebrovascular Disorders/surgery , Chemokine CCL11/metabolism , Chemokines, CC/metabolism , Disease Models, Animal , Hypoxia-Ischemia, Brain/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Primary Cell Culture , Receptors, CCR3/antagonists & inhibitors , Receptors, CCR3/genetics , Receptors, CCR3/metabolism
11.
Stem Cells Transl Med ; 6(3): 713-719, 2017 03.
Article in English | MEDLINE | ID: mdl-28297565

ABSTRACT

Mesenchymal stem (stromal) cells (MSCs) are being investigated for treating degenerative and inflammatory disorders because of their reparative and immunomodulatory properties. Intricate mechanisms relate cell death processes with immune responses, which have implications for degenerative and inflammatory conditions. We review the therapeutic value of MSCs in terms of preventing regulated cell death (RCD). When cells identify an insult, specific intracellular pathways are elicited for execution of RCD processes, such as apoptosis, necroptosis, and pyroptosis. To some extent, exacerbated RCD can provoke an intense inflammatory response and vice versa. Emerging studies are focusing on the molecular mechanisms deployed by MSCs to ameliorate the survival, bioenergetics, and functions of unfit immune or nonimmune cells. Given these aspects, and in light of MSC actions in modulating cell death processes, we suggest the use of novel functional in vitro assays to ensure the potency of MSCs for preventing RCD. Such analyses should be associated with existing functional assays measuring the anti-inflammatory capabilities of MSCs in vitro. MSCs selected on the basis of two in vitro functional criteria (i.e., prevention of inflammation and RCD) could possess optimal therapeutic efficacy in vivo. In addition, we underline the implications of these perspectives in clinical studies of MSC therapy, with particular focus on acute respiratory distress syndrome. Stem Cells Translational Medicine 2017;6:713-719.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Death , Humans
12.
Sci Rep ; 6: 26162, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27194621

ABSTRACT

The biological effects of indium-tin-oxide (ITO) are of considerable importance because workers exposed to indium compounds have been diagnosed with interstitial lung disease or pulmonary alveolar proteinosis; however, the pathophysiology of these diseases is undefined. Here, mice intraperitoneally inoculated with ITO-nanoparticles (ITO-NPs) resulted in peritonitis dependent in NLRP3 inflammasome, with neutrophils recruitment and interleukin-1ß (IL-1ß) production. Withal peritoneal macrophages exposed ex vivo to ITO-NPs caused IL-1ß secretion and cytolysis. Further, alveolar macrophages exposed to ITO-NPs in vitro showed ITO-NP endocytosis and production of tumor necrosis factor-α (TNF-α) and IL-1ß, ensued cell death by cytolysis. This cell death was RIPK1-independent but caspase1-dependent, and thus identified as pyroptosis. Endocytosis of ITO-NPs by activated THP-1 cells induced pyroptosis with IL-1ß/TNF-α production and cytolysis, but not in activated THP-1 cells with knockdown of NLRP3, ASC, or caspase1. However, exposing activated THP-1 cells with NLRP3 or ASC knockdown to ITO-NPs resulted in cell death but without cytolysis, with deficiency in IL-1ß/TNF-α, and revealing features of apoptosis. While, mesenchymal stem cells (MSCs) co-cultured with macrophages impaired both inflammation and cell death induced by ITO-NPs. Together, our findings provide crucial insights to the pathophysiology of respiratory diseases caused by ITO particles, and identify MSCs as a potent therapeutic.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Macrophages/metabolism , Mesenchymal Stem Cells/physiology , Pyroptosis , Receptors, Cell Surface/metabolism , Tin Compounds/metabolism , Animals , Cells, Cultured , Coculture Techniques , Endocytosis , Humans , Interleukin-1beta/metabolism , Mice , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Neutrophils/immunology , Peritonitis/chemically induced , Tin Compounds/administration & dosage
13.
J Matern Fetal Neonatal Med ; 28(7): 842-7, 2015 May.
Article in English | MEDLINE | ID: mdl-24939627

ABSTRACT

OBJECTIVE: Neonatal hypoxic-ischemic encephalopathy (HIE) is caused by brain injury that occurs in a developing fetus or infant. Stem cell transplantation can reportedly induce functional recovery in animal models of HIE. Murine neonatal splenocytes are enriched with immature blood stem cells and are used for the investigation of murine models of syngeneic transplantation. The aim of this study was to investigate the therapeutic potential of newborn splenocytes in a murine model of neonatal ischemia-reperfusion brain injury. METHODS: C57BL/6N mice (postnatal day 7) underwent right common carotid artery occlusion with an aneurysm clip. Following hypoxic exposure, reperfusion was achieved by unclamping the artery. Newborn splenocytes were transplanted intravenously at 3 weeks after injury. RESULTS: The splenocytes transplanted group tended to show an improvement in behavioral tests, but it was not significantly different compared with the control groups. The transplanted cells were localized in various organs including injured brain tissue over 3 weeks. In the penumbra region of the brain, vascular endothelial growth factor (VEGF) expression was upregulated after transplantation. CONCLUSIONS: These results showed that syngeneic transplantation of newborn splenocytes achieved the long-term survival of the grafts and exerted influence the microenvironment in the injured brains of mice.


Subject(s)
Hypoxia-Ischemia, Brain/therapy , Reperfusion Injury/therapy , Spleen/cytology , Stem Cell Transplantation , Animals , Blotting, Western , Flow Cytometry , Hypoxia-Ischemia, Brain/diagnosis , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Reperfusion Injury/diagnosis , Transplantation, Isogeneic , Treatment Outcome
14.
J Leukoc Biol ; 98(4): 671-81, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26162403

ABSTRACT

Human Slan DCs have been studied in patients with psoriasis, rheumatoid arthritis, cancer, and autoimmune diseases. In this study, we investigated the frequency, phenotype, and function of Slan DCs in blood, colon, as well as mLNs of patients with IBD. We first show that the frequency of circulating CD14(dull)Slan DCs was reduced in CD patients refractory to immunosuppressive drugs or TNF-α blockers relative to untreated CD, UC, and healthy subjects. In blood of CD patients, Slan DCs expressed CD172a, as detected by CD47 fusion protein binding, when compared with its lack of expression in control subjects. Next, we demonstrate that CD172a(+)Slan DCs that produced IL-1ß and TNF-α accumulated in mLNs and colons of CD patients. The CD172a(+)Slan DCs up-regulated their expression of CD14 in CD tissues and the proinflammatory cytokines were produced in CD14(bright)CD172a(+)Slan DCs. By contrast, no difference was noted in the frequency of Slan DCs between inflamed, noninflamed colonic mucosa of UC patients and control, non-IBD donors. Finally, the percentage of cytokine-producing Slan DCs also augmented in response to TLR2 and NOD2 in in vitro stimulation in PBMCs of CD, but not UC, patients. In conclusion, we propose that proinflammatory CD14(bright)CD172a(+)Slan DCs are a distinguishing feature between CD and UC, as these cells accumulate uniquely in mLNs and colonic mucosa of CD patients. Thus, Slan DCs may contribute to CD immunopathogenesis.


Subject(s)
Colitis, Ulcerative/immunology , Colon/immunology , Crohn Disease/immunology , Dendritic Cells/immunology , Lymph Nodes/immunology , Adult , Aged , Amino Sugars/biosynthesis , Amino Sugars/immunology , Cytokines/biosynthesis , Female , Flow Cytometry , Humans , Immunohistochemistry , Intestinal Mucosa/immunology , Male , Middle Aged , Young Adult
15.
J Exp Med ; 210(6): 1251-63, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23669395

ABSTRACT

In mice, the transfer of CD172a(+) (SIRP-α) dendritic cells (DCs) elicits T cell-driven colitis, whereas treatment with CD47-Fc protein, a CD172a-binding agent, confers protection. The aim of this study was to elucidate the nature and functional properties of human CD172a(+) DCs in chronic intestinal inflammation. Here, we show that CD172a(+)CD11c(+) cells accumulate in the mesenteric lymph nodes (mLNs) and inflamed intestinal mucosa in patients with Crohn's disease (CD). These cells are distinct from resident DCs and may coexpress markers typically associated with monocyte-derived inflammatory DCs such as CD14 and/or DC-SIGN, E-Cadherin, and/or CX3CR1. Spontaneous IL-1ß and TNF production by HLA-DR(+) cells in CD tissues is restricted to those expressing CD172a. An avidity-improved CD47 fusion protein (CD47-Var1) suppresses the release of a wide array of inflammatory cytokines by CD172a(+) cells, which may include HLA-DR(-)CD172a(+) neutrophils, in inflamed colonic explant cultures and impairs the ability of HLA-DR(+)CD172a(+) cells to activate memory Th17 but not Th1 responses in mLNs. In conclusion, targeting CD172a(+) cells may represent novel therapeutic perspectives for patients with CD.


Subject(s)
Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , Crohn Disease/immunology , Interleukin-1beta/metabolism , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , CX3C Chemokine Receptor 1 , Cadherins/metabolism , Dendritic Cells/metabolism , HLA-DR Antigens/metabolism , Humans , Inflammation/metabolism , Intestinal Mucosa/metabolism , Lymph Nodes/metabolism , Monocytes/metabolism , Receptors, Chemokine/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism
16.
Hum Immunol ; 73(8): 795-800, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22609446

ABSTRACT

The low molecular weight compound VAF347, and its pro-drug version VAG539, interact with the transcription factor aryl hydrocarbon receptor (AhR) on monocytes to mediate its anti-inflammatory activity in vitro and in vivo. AhR is a crucial factor for IL-22 production, which regulates skin and gut homeostasis. Here we investigated whether VAF347 might control the differentiation of naïve T cells into IL-22-secreting cells and/or regulate IL-22 production by memory T cells. Human monocytes exposed to VAF347 differentiated into dendritic cells capable of instructing a naïve CD4(+) T cell differentiation program that promoted IL-22 secretion and concomitantly inhibited IL-17 production. Whilst AhR ligation by VAF347 on naïve CD4(+) T cells favored the development of single IL-22-secreting cells (Th22), it suppressed the generation of T cells secreting either IL-22 and IFN-γ or IL-17 and IFN-γ. In contrast, memory T cells were refractory to AhR regulation since VAF347, AhR antagonist or AhR gene silencing did not modulate the production of any of these cytokines. Interfering with AhR functions using VAF347 may provide an efficient way to intervene with autoimmune disease since it would enhance the host protective function mediated by IL-22 while preventing the development of Th cells secreting pro-inflammatory cytokines.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Dendritic Cells/drug effects , Interleukins/immunology , Monocytes/drug effects , Pyrimidines/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immunologic Memory , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukins/biosynthesis , Interleukins/metabolism , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/metabolism , Prodrugs/metabolism , Prodrugs/pharmacology , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction/immunology , Interleukin-22
17.
PLoS One ; 7(8): e41972, 2012.
Article in English | MEDLINE | ID: mdl-22870271

ABSTRACT

How do effector CD4 T cells escape cell death during the contraction of the immune response (IR) remain largely unknown. CD47, through interactions with thrombospondin-1 (TSP-1) and SIRP-α, is implicated in cell death and phagocytosis of malignant cells. Here, we reported a reduction in SIRP-α-Fc binding to effector memory T cells (T(EM)) and in vitro TCR-activated human CD4 T cells that was linked to TSP-1/CD47-induced cell death. The reduced SIRP-α-Fc binding (CD47(low) status) was not detected when CD4 T cells were stained with two anti-CD47 mAbs, which recognize distinct epitopes. In contrast, increased SIRP-α-Fc binding (CD47(high) status) marked central memory T cells (T(CM)) as well as activated CD4 T cells exposed to IL-2, and correlated with resistance to TSP-1/CD47-mediated killing. Auto-aggressive CD4 effectors, which accumulated in lymph nodes and at mucosal sites of patients with Crohn's disease, displayed a CD47(high) status despite a high level of TSP-1 release in colonic tissues. In mice, CD47 (CD47(low) status) was required on antigen (Ag)-specific CD4 effectors for the contraction of the IR in vivo, as significantly lower numbers of Ag-specific CD47(+/+)CD4 T cells were recovered when compared to Ag-specific CD47(-/-) CD4 T cells. In conclusion, we demonstrate that a transient change in the status of CD47, i.e. from CD47(high) to CD47(low), on CD4 effectors regulates the decision-making process that leads to CD47-mediated cell death and contraction of the IR while maintenance of a CD47(high) status on tissue-destructive CD4 effectors prevents the resolution of the inflammatory response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD47 Antigen/immunology , Clonal Deletion , Immunologic Memory , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , CD4-Positive T-Lymphocytes/pathology , CD47 Antigen/genetics , Cell Death/genetics , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Female , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Male , Mice , Mice, Knockout , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology
18.
PLoS One ; 4(3): e5087, 2009.
Article in English | MEDLINE | ID: mdl-19333372

ABSTRACT

Th17 cells are implicated in host defence and autoimmune diseases. CD28/B7 co-stimulation is involved in the induction and progression of autoimmune diseases, but its role in controlling murine Th17 cell fate remains to be clarified. We here report that soluble anti-CD28 mAb suppressed the differentiation of anti-CD3-stimulated naïve CD4(+) T cells into IL-17-producing cells. CD28 co-stimulation reduced the frequency of proliferating cells that produce IL-17. We provide evidence for an IL-2 and IFN-gamma-dependent mechanism of CD28-mediated IL-17 suppression. CD28 blockade of Th17 development was correlated with a decrease rather than an increase in the percentage of Foxp3(+) T cells. In APC/T cell co-cultures, mature dendritic cells (DC) were less efficient than immature DC in their ability to support Th17 cell differentiation, while CTLA4-Ig, an agent blocking CD28/B7 and CTLA4/B7 interactions, facilitated both murine and human Th17 differentiation. This study identifies the importance of B7 co-stimulatory molecules in the negative regulation of Th17 development. These unexpected results caution targeting the CD28/B7 pathways in the treatment of human autoimmune diseases.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , Interleukin-17/biosynthesis , Lymphocyte Activation , Animals , Coculture Techniques , Dendritic Cells , Down-Regulation/physiology , Humans , Interferon-gamma , Interleukin-2 , Mice , T-Lymphocyte Subsets
19.
Nat Genet ; 41(1): 71-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19098911

ABSTRACT

We used a candidate gene approach to identify a set of SNPs, located in a predicted regulatory region on chromosome 1q44 downstream of NLRP3 (previously known as CIAS1 and NALP3) that are associated with Crohn's disease. The associations were consistently replicated in four sample sets from individuals of European descent. In the combined analysis of all samples (710 father-mother-child trios, 239 cases and 107 controls), these SNPs were strongly associated with risk of Crohn's disease (P(combined) = 3.49 x 10(-9), odds ratio = 1.78, confidence interval = 1.47-2.16 for rs10733113), reaching a level consistent with the stringent significance thresholds imposed by whole-genome association studies. In addition, we observed significant associations between SNPs in the associated regions and NLRP3 expression and IL-1beta production. Mutations in NLRP3 are known to be responsible for three rare autoinflammatory disorders. These results suggest that the NLRP3 region is also implicated in the susceptibility of more common inflammatory diseases such as Crohn's disease.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Base Pairing , Carrier Proteins/metabolism , Case-Control Studies , Gene Expression Regulation , Humans , NLR Family, Pyrin Domain-Containing 3 Protein , Reproducibility of Results
20.
J Allergy Clin Immunol ; 118(6): 1342-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17137867

ABSTRACT

BACKGROUND: Analysis of naturally occurring T regulatory CD4+ (nTreg) cells in human diseases is hampered by the lack of specific surface marker. Indeed, the CD25 antigen, which is typically used to identify nTreg cells, is also expressed on activated effector T cells. OBJECTIVE: We sought to examine whether CD4+ T cells bearing CD103 are suppressor cells, regardless of CD25 coexpression. METHODS: We first compared freshly isolated tonsillar CD103+ CD25- cells with their CD103- CD25high counterparts for their capacity to suppress T-cell response and their expression of FoxP3 mRNA. Next CD103 was induced on neonatal or adult CD4+ T cells stimulated with allogeneic dendritic cells, and the CD103+ and CD103- fractions were compared as above. RESULTS: Tonsillar CD4+ CD103+ CD25- T cells displayed comparable suppressive activity and contained similar amounts of FoxP3 mRNA as their CD103- CD25high counterparts. In vitro-generated alloantigen-primed CD103+ cells coexpressed CD25, suppressed T-cell activation, and contained more FoxP3 mRNA than the CD103- CD25+ cells isolated from the same cultures. Finally, neonatal alloreactive cells contained more CD103+ Treg cells than their adult counterparts and, unlike the latter, became hyporesponsive to the priming alloantigens. CONCLUSIONS: The examination of CD103 and CD25 coexpression allows identification of 3 subsets of human CD4+ nTreg cells, and the detection of CD103 on CD4+ T cells identifies nTreg cells, regardless of CD25 coexpression. CLINICAL IMPLICATIONS: The greater induction of CD103+ suppressor cells by cord blood should be related to its successful clinical use as an alternative to adult bone marrow transplantation.


Subject(s)
Antigens, CD/biosynthesis , Integrin alpha Chains/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Age Factors , Antigens, CD/immunology , Biomarkers , Cells, Cultured , Coculture Techniques , Humans , Integrin alpha Chains/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , T-Lymphocyte Subsets/classification
SELECTION OF CITATIONS
SEARCH DETAIL