Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Stem Cells Transl Med ; 12(4): 194-206, 2023 04 17.
Article in English | MEDLINE | ID: mdl-36928395

ABSTRACT

Mesenchymal stromal cells (MSCs) have recently emerged as an interesting therapeutic approach for patients with progressive systemic sclerosis (SSc), a rare and life-threatening orphan autoimmune disease. Whereas MSC immunomodulatory potential is considered as a central mechanism for their clinical benefit, very few data are available on the impact of MSCs on immune cell subsets in vivo. In the current extended study of a phase I/II clinical trial exploring the injection of a single dose of allogeneic bone marrow-MSCs (alloBM-MSCs) in patients with severe SSc (NCT02213705), we performed a longitudinal in-depth characterization of circulating immune cells in 19 MSC-treated patients, including 14 responders and 5 non-responders. By a combination of flow cytometry and transcriptomic analyses, we highlighted an increase in circulating CD24hiCD27posCD38lo/neg memory B cells, the main IL-10-producing regulatory B cell (Breg) subset, and an upregulation of IL10 expression in ex-vivo purified B cells, specifically in responder patients, early after the alloBM-MSC infusion. In addition, a deeper alteration of the B-cell compartment before alloBM-MSC treatment, including a higher expression of profibrotic cytokines IL6 and TGFß by sorted B cells was associated with a non-responder clinical status. Finally, BM-MSCs were able to directly upregulate IL-10 production in activated B cells in vitro. These data suggest that cytokine-producing B cells, in particular Breg, are pivotal effectors of BM-MSC therapeutic activity in SSc. Their quantification as activity biomarkers in MSC potency assays and patient selection criteria may be considered to reach optimal clinical benefit when designing MSC-based clinical trials.


Subject(s)
B-Lymphocytes, Regulatory , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Scleroderma, Systemic , Humans , Interleukin-10/metabolism , Bone Marrow , Cytokines/metabolism , Scleroderma, Systemic/therapy , Scleroderma, Systemic/metabolism
2.
Cytotherapy ; 24(5): 500-507, 2022 05.
Article in English | MEDLINE | ID: mdl-35219585

ABSTRACT

The therapeutic potential of culture-adapted adipose-derived stromal cells (ASCs) is largely related to their production of immunosuppressive factors that are inducible in vitro by priming with inflammatory stimuli, in particular tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ). In vivo, obesity is associated with chronic inflammation of white adipose tissue, including accumulation of neutrophils, infiltration by IFNγ/TNFα-producing immune cells, and ASC dysfunction. In the current study, we identified in obese patients a simultaneous upregulation of CD40Lin the adipose tissue stroma vascular fraction (AT-SVF), correlated with the Th1 gene signature, and an overexpression of CD40 by native ASCs. Moreover, activated CD4+ T cells upregulated CD40 on culture-expanded ASCs and triggered their production of IL-8 in a CD40L-dependent manner, leading to an increased capacity to recruit neutrophils. Finally, activation of ASCs by sCD40L or CD40L-expressing CD4+ T cells relies on both canonical and non-canonical NF-κB pathways, and IL-8 was found to be coregulated with NF-κB family members in AT-SVF. These data identify the CD40-CD40L axis as a priming mechanism of ASCs, able to modulate their cross talk with neutrophils in an inflammatory context, and their functional capacity for therapeutic applications.


Subject(s)
CD40 Ligand , NF-kappa B , Adipose Tissue , CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/genetics , CD40 Ligand/metabolism , Cells, Cultured , Chemokines/metabolism , Humans , Interleukin-8/metabolism , NF-kappa B/metabolism , Obesity , Stromal Cells/pathology , T-Lymphocytes , Tumor Necrosis Factor-alpha/metabolism
3.
Cell Rep Med ; 2(6): 100291, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33977279

ABSTRACT

Acute respiratory distress syndrome (ARDS) is the main complication of coronavirus disease 2019 (COVID-19), requiring admission to the intensive care unit (ICU). Despite extensive immune profiling of COVID-19 patients, to what extent COVID-19-associated ARDS differs from other causes of ARDS remains unknown. To address this question, here, we build 3 cohorts of patients categorized in COVID-19-ARDS+, COVID-19+ARDS+, and COVID-19+ARDS-, and compare, by high-dimensional mass cytometry, their immune landscape. A cell signature associating S100A9/calprotectin-producing CD169+ monocytes, plasmablasts, and Th1 cells is found in COVID-19+ARDS+, unlike COVID-19-ARDS+ patients. Moreover, this signature is essentially shared with COVID-19+ARDS- patients, suggesting that severe COVID-19 patients, whether or not they experience ARDS, display similar immune profiles. We show an increase in CD14+HLA-DRlow and CD14lowCD16+ monocytes correlating to the occurrence of adverse events during the ICU stay. We demonstrate that COVID-19-associated ARDS displays a specific immune profile and may benefit from personalized therapy in addition to standard ARDS management.


Subject(s)
COVID-19/pathology , Leukocytes, Mononuclear/metabolism , Respiratory Distress Syndrome/immunology , Aged , COVID-19/complications , COVID-19/virology , Cohort Studies , Evolution, Molecular , Female , HLA-DR Antigens/metabolism , Humans , Intensive Care Units , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lipopolysaccharide Receptors/metabolism , Machine Learning , Male , Middle Aged , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , SARS-CoV-2/isolation & purification , Sialic Acid Binding Ig-like Lectin 1/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism
4.
Front Immunol ; 12: 653577, 2021.
Article in English | MEDLINE | ID: mdl-34017332

ABSTRACT

Multiple sclerosis (MS) is an immune-driven demyelinating disease of the central nervous system. Immune cell features are particularly promising as predictive biomarkers due to their central role in the pathogenesis but also as drug targets, even if nowadays, they have no impact in clinical practice. Recently, high-resolution approaches, such as mass cytometry (CyTOF), helped to better understand the diversity and functions of the immune system. In this study, we performed an exploratory analysis of blood immune response profiles in healthy controls and MS patients sampled at their first neurological relapse, using two large CyTOF panels including 62 markers exploring myeloid and lymphoid cells. An increased abundance of both a T-bet-expressing B cell subset and a CD206+ classical monocyte subset was detected in the blood of early MS patients. Moreover, T-bet-expressing B cells tended to be enriched in aggressive MS patients. This study provides new insights into understanding the pathophysiology of MS and the identification of immunological biomarkers. Further studies will be required to validate these results and to determine the exact role of the identified clusters in neuroinflammation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Monocytes/immunology , Multiple Sclerosis/immunology , Adult , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , Biomarkers/blood , Cell Separation/methods , Cohort Studies , Female , Flow Cytometry/methods , Humans , Male , Membrane Glycoproteins/metabolism , Monocytes/metabolism , Multiple Sclerosis/blood , Receptors, Immunologic/metabolism , T-Box Domain Proteins/metabolism , Young Adult
5.
Blood Adv ; 5(8): 2063-2074, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33877296

ABSTRACT

The immunomodulatory drug lenalidomide is used in patients with follicular lymphoma (FL) with the aim of stimulating T-cell antitumor immune response. However, little is known about the effects of lenalidomide on T-cell biology in vivo in patients with FL. We thus undertook an extensive longitudinal immunologic study, including phenotypic, transcriptomic, and functional analyses, on 44 first-line and 27 relapsed/refractory patients enrolled in the GALEN trial (Obinutuzumab Combined With Lenalidomide for Relapsed or Refractory Follicular B-Cell Lymphoma) to test the efficacy of lenalidomide and obinutuzumab combination in patients with FL. Lenalidomide rapidly and transiently induced an activated T-cell phenotype, including HLA-DR, Tim-3, CD137, and programmed cell death protein 1 (PD-1) upregulation. Furthermore, sequential RNA-sequencing of sorted PD-1+ and PD-1- T-cell subsets revealed that lenalidomide triggered a strong enrichment for several gene signatures related to effector memory T-cell features, including proliferation, antigen receptor signaling, and immune synapse restoration; all were validated at the phenotypic level and with ex vivo functional assays. Correlative analyses pinpointed a negative clinical impact of high effector T-cell and regulatory T-cell percentages before and during treatment. Our findings bring new insight in lenalidomide mechanisms of action at work in vivo and will fuel a new rationale for the design of combination therapies.


Subject(s)
Lymphoma, B-Cell , Lymphoma, Follicular , Humans , Immunomodulation , Lenalidomide/therapeutic use , Lymphoma, Follicular/drug therapy
6.
Stem Cells ; 38(1): 146-159, 2020 01.
Article in English | MEDLINE | ID: mdl-31502731

ABSTRACT

Clinical-grade mesenchymal stromal cells (MSCs) can be expanded from bone marrow and adipose tissue to treat inflammatory diseases and degenerative disorders. However, the influence of their tissue of origin on their functional properties, including their immunosuppressive activity, remains unsolved. In this study, we produced paired bone marrow-derived mesenchymal stromal cell (BM-MSC) and adipose-derived stromal cell (ASC) batches from 14 healthy donors. We then compared them using transcriptomic, phenotypic, and functional analyses and validated our results on purified native MSCs to infer which differences were really endowed by tissue of origin. Cultured MSCs segregated together owing to their tissue of origin based on their gene expression profile analyzed using differential expression and weighted gene coexpression network analysis. This translated into distinct immune-related gene signatures, phenotypes, and functional cell interactions. Importantly, sorted native BM-MSCs and ASCs essentially displayed the same distinctive patterns than their in vitro-expanded counterparts. As a whole, ASCs exhibited an immune profile consistent with a stronger inhibition of immune response and a lower immunogenicity, supporting the use of adipose tissue as a valuable source for clinical applications.


Subject(s)
Mesenchymal Stem Cells/metabolism , Transcriptome/genetics , Adult , Cell Differentiation , Cells, Cultured , Female , Humans , Male , Middle Aged , Phenotype , Young Adult
7.
Stem Cells ; 35(5): 1431-1436, 2017 05.
Article in English | MEDLINE | ID: mdl-28142215

ABSTRACT

Owing to their immunosuppressive properties, mesenchymal stromal cells (MSCs) obtained from bone marrow (BM-MSCs) or adipose tissue (ASCs) are considered a promising tool for cell therapy. However, important issues should be considered to ensure the reproducible production of efficient and safe clinical-grade MSCs. In particular, high expansion rate, associated with progressive senescence, was recently proposed as one of the parameters that could alter MSC functionality. In this study, we directly address the consequences of replicative senescence on BM-MSC and ASC immunomodulatory properties. We demonstrate that MSCs produced according to GMP procedures inhibit less efficiently T-cell, but not Natural Killer (NK)- and B-cell, proliferation after reaching senescence. Senescence-related loss-of-function is associated with a decreased indoleamine 2,3-dioxygenase (IDO) activity in response to inflammatory stimuli. In particular, although STAT-1-dependent IDO expression is transcriptionally induced at a similar level in senescent and nonsenescent MSCs, IDO protein is specifically degraded by the proteasome in senescent ASCs and BM-MSCs, a process that could be reversed by the MG132 proteasome inhibitor. These data encourage the use of appropriate quality controls focusing on immunosuppressive mechanisms before translating clinical-grade MSCs in the clinic. Stem Cells 2017;35:1431-1436.


Subject(s)
Cellular Senescence , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Cell Proliferation , Humans , T-Lymphocytes/cytology
8.
J Infect Dis ; 201(6): 956-66, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20151841

ABSTRACT

BACKGROUND: Severe sepsis results in a sustained deleterious immune dysregulation. Indoleamine 2,3-dioxygenase (IDO), the rate-limiting enzyme of tryptophan catabolism, plays a pivotal role in immune tolerance and is induced during various inflammatory conditions. METHODS: Plasma samples obtained from patients with septic shock (n = 38), severe sepsis (n = 35), or sepsis (n = 10) and from healthy donors (n = 26) were analyzed for IDO activity by high-performance liquid chromatography. Lymphocyte, monocyte, and regulatory T cell counts as well as monocytic human leukocyte antigen DR (HLA-DR) expression were quantified by flow cytometry. Peripheral blood mononuclear cells and purified CD14(+) and CD14(-) fractions were assayed in vitro for spontaneous and inducible IDO expression and activity. RESULTS: IDO activity gradually increased according to sepsis severity, and septic patients who died had higher IDO activity on admission than did survivors (P = .013). Monocytes were a major source of active IDO in normal peripheral blood. The percentage and absolute number of circulating CD14(+) cells were increased in septic patients, and their monocytes remained fully able to produce functional IDO after NF-kappaB-independent interferon gamma stimulation but not through NF-kappaB-dependent Toll-like receptor engagement. CONCLUSIONS: IDO activity is increased during severe sepsis and septic shock and is associated with mortality. IDO production could be used to better characterize monocyte reprogramming in sepsis.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Sepsis/enzymology , Adult , Aged , CD4 Lymphocyte Count , Case-Control Studies , Chromatography, High Pressure Liquid , Female , Flow Cytometry , France/epidemiology , HLA-DR Antigens/blood , Humans , Male , Middle Aged , NF-kappa B/physiology , Sepsis/blood , Sepsis/mortality , Shock, Septic/blood , Shock, Septic/enzymology , Shock, Septic/mortality , T-Lymphocytes, Regulatory , Young Adult
9.
Cancer Res ; 69(7): 3228-37, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276371

ABSTRACT

Human mesenchymal stem cells (MSC) strongly repress activated T-cell proliferation through the production of a complex set of soluble factors, including the tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO), which is induced by IFN-gamma. Conversely, MSCs support survival of follicular lymphoma (FL) B cells, in particular after exposure to tumor necrosis factor-alpha (TNF) and lymphotoxin-alpha1beta2 (LT). The role of MSCs on normal and malignant B-cell growth in steady-state and inflammatory conditions remains to be fully explored. We show here that resting MSCs sustain activated normal B-cell proliferation and survival, whereas IFN-gamma-conditioned MSCs mediate IDO-dependent B-cell growth arrest and apoptosis. IFN-gamma, TNF, and LT are significantly overexpressed by the microenvironment of invaded FL-lymph nodes, but their relative expression patterns are highly heterogeneous between samples. In vitro, IFN-gamma abrogates the B-cell supportive phenotype induced by TNF and LT on MSCs. Moreover, IFN-gamma overrules the growth promoting effect of MSCs on primary purified FL B cells. Altogether, these results underline the crucial role of the cytokine context in the local crosstalk between malignant cells and their microenvironment and provide new insights into our knowledge of the FL cell niche that emerges as a new promising target for innovative therapeutic strategies.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/pharmacology , Lymphoma, Follicular/enzymology , Lymphoma, Follicular/pathology , Mesoderm/enzymology , Mesoderm/pathology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Survival/immunology , Child , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-gamma/immunology , Lymphocyte Activation , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/immunology , Mesoderm/immunology , Stromal Cells/enzymology , Stromal Cells/immunology , Stromal Cells/pathology
10.
Crit Care Med ; 34(4): 1193-201, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16484916

ABSTRACT

OBJECTIVE: Loss of surface human leukocyte antigen-DR (HLA-DR) on monocytes is a major factor of immunosuppression in sepsis. Granulocyte macrophage-colony stimulating factor (GM-CSF) up-regulates HLA-DR expression on monocytes via the GM-CSF receptor (GM-CSFr) through a transcriptional mechanism involving the class II transactivator factor (CIITA). We investigated monocyte GM-CSFr expression and its relationship with HLA-DR in septic patients. DESIGN: Prospective clinical experimental study. SETTING: University hospital intensive care unit and research facility. PATIENTS: Septic patients with and without septic shock, control patients. INTERVENTIONS: Flow cytometry and real-time quantitative reverse polymerase chain reaction were used to characterize GM-CSFr expression and transcription in septic patients and in ex vivo stimulated healthy monocytes. MEASUREMENTS AND MAIN RESULTS: We showed an early GM-CSFr down-modulation in patients with septic shock compared with those without septic shock and controls. A persistent low GM-CSFr expression was observed in patients who acquired secondary infections or in those who died, and this persistent defect correlated with severity scores. We demonstrated that GM-CSFr down-modulation occurs at a posttranscriptional level since we observed no alteration in GM-CSFr transcription in monocytes isolated from septic patients. Furthermore, we demonstrated that GM-CSFr expression levels on monocytes correlated not only with HLA-DR expression and transcription levels but also with RNA levels of its main transcriptional factor CIITA. Because we previously showed in septic patients a relationship between high cortisol plasma level and low monocyte HLA-DR expression, we investigated the effects of glucocorticoids on monocyte GM-CSFr expression and observed a similar posttranscriptional down-modulation of GM-CSFr by steroids. However, the in vivo putative role of steroids in HLA-DR down-regulation via GM-CSFr down-modulation needs further investigation. CONCLUSION: Monocyte GM-CSFr down-modulation occurred in septic shock, was associated with severity, and might be either another manifestation of monocyte deactivation linked to sepsis or an additional mechanism participating in immunosuppression.


Subject(s)
Monocytes/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Shock, Septic/immunology , Adult , Aged , Down-Regulation , Humans , Middle Aged , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...