Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
2.
Blood ; 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38643512

ABSTRACT

Plasma cells (PC) are highly specialized cells representing the end stage of B cell differentiation. We have shown that PC differentiation can be reproduced in vitro using elaborate culture systems. The molecular changes occurring during PC differentiation are recapitulated in this in vitro differentiation model. However, a major challenge exists to decipher the spatiotemporal epigenetic and transcriptional programs that drives the early stages of PC differentiation. We combined single cell (sc) RNA-seq and single cell ATAC-seq to decipher the trajectories involved in PC differentiation. ScRNA-seq experiments revealed a strong heterogeneity of the preplasmablastic and plasmablastic stages. Among genes that were commonly identified using scATAC-seq and scRNA-seq, we identified several transcription factors with significant stage specific potential importance in PC differentiation. Interestingly, differentially accessible peaks characterizing the preplasmablastic stage were enriched in motifs of BATF3, FOS and BATF, belonging to the AP-1 transcription factor family, that may represent key transcriptional nodes involved in PCD. Integration of transcriptomic and epigenetic data at the single cell level revealed that a population of preplasmablasts already undergone epigenetic remodeling related to PC profile together with UPR activation and are committed to differentiate in PC. These results and the supporting data generated with our in vitro PC differentiation model provide a unique resource for the identification of molecular circuits that are crucial for early and mature plasma cell maturation and biological functions. These data thus provide critical insights into epigenetic- and transcriptional-mediated reprogramming events that sustain PC differentiation.

3.
Br J Haematol ; 204(4): 1383-1392, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38442908

ABSTRACT

Warts, hypogammaglobulinaemia, infections and myelokathexis syndrome (WHIMS) is a rare combined primary immunodeficiency caused by the gain of function of the CXCR4 chemokine receptor. We present the prevalence of cancer in WHIMS patients based on data from the French Severe Chronic Neutropenia Registry and an exhaustive literature review. The median follow-up of the 14 WHIMS 'patients was 28.5 years. A central review and viral evaluation of pathological samples were organized, and we conducted a thorough literature review to identify all reports of WHIMS cases. Six French patients were diagnosed with cancer at a median age of 37.6 years. The 40-year risk of malignancy was 39% (95% confidence interval [CI]: 6%-74%). We observed two human papillomavirus (HPV)-induced vulvar carcinomas, three lymphomas (two Epstein-Barr virus [EBV]-related) and one basal cell carcinoma. Among the 155 WHIMS cases from the literature, 22 cancers were reported in 16 patients, with an overall cancer 40-year risk of 23% (95% CI: 13%-39%). Malignancies included EBV-associated lymphoproliferative disorders and HPV-positive genital and anal cancers as in the French cohort. Worldwide, nine cases of malignancy were associated with HPV and four with EBV. Immunocompromised WHIMS patients appear to be particularly susceptible to developing early malignancy, mainly HPV-induced carcinomas, followed by EBV-related lymphomas.


Subject(s)
Agammaglobulinemia , Carcinoma , Epstein-Barr Virus Infections , Lymphoma , Papillomavirus Infections , Primary Immunodeficiency Diseases , Warts , Humans , Adult , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/epidemiology , Herpesvirus 4, Human , Warts/complications , Warts/epidemiology , Warts/diagnosis , Syndrome , Receptors, CXCR4
6.
Eur J Immunol ; 53(9): e2250334, 2023 09.
Article in English | MEDLINE | ID: mdl-37377335

ABSTRACT

Bone marrow (BM) long-lived plasma cells (PCs) are essential for long-term protection against infection, and their persistence within this organ relies on interactions with Cxcl12-expressing stromal cells that are still not clearly identified. Here, using single cell RNAseq and in silico transinteractome analyses, we identified Leptin receptor positive (LepR+ ) mesenchymal cells as the stromal cell subset most likely to interact with PCs within the BM. Moreover, we demonstrated that depending on the isotype they express, PCs may use different sets of integrins and adhesion molecules to interact with these stromal cells. Altogether, our results constitute an unprecedented characterization of PC subset stromal niches and open new avenues for the specific targeting of BM PCs based on their isotype.


Subject(s)
Bone Marrow , Mesenchymal Stem Cells , Bone Marrow/metabolism , Plasma Cells , Stromal Cells , Cell Adhesion Molecules/metabolism , Bone Marrow Cells
7.
Nat Immunol ; 24(7): 1124-1137, 2023 07.
Article in English | MEDLINE | ID: mdl-37217705

ABSTRACT

The magnitude and quality of the germinal center (GC) response decline with age, resulting in poor vaccine-induced immunity in older individuals. A functional GC requires the co-ordination of multiple cell types across time and space, in particular across its two functionally distinct compartments: the light and dark zones. In aged mice, there is CXCR4-mediated mislocalization of T follicular helper (TFH) cells to the dark zone and a compressed network of follicular dendritic cells (FDCs) in the light zone. Here we show that TFH cell localization is critical for the quality of the antibody response and for the expansion of the FDC network upon immunization. The smaller GC and compressed FDC network in aged mice were corrected by provision of TFH cells that colocalize with FDCs using CXCR5. This demonstrates that the age-dependent defects in the GC response are reversible and shows that TFH cells support stromal cell responses to vaccines.


Subject(s)
T-Lymphocytes, Helper-Inducer , Vaccines , Animals , Mice , B-Lymphocytes , T Follicular Helper Cells , Germinal Center , Aging
8.
Nat Commun ; 14(1): 2058, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37045841

ABSTRACT

WHIM Syndrome is a rare immunodeficiency caused by gain-of-function CXCR4 mutations. Here we report a decrease in bone mineral density in 25% of WHIM patients and bone defects leading to osteoporosis in a WHIM mouse model. Imbalanced bone tissue is observed in mutant mice combining reduced osteoprogenitor cells and increased osteoclast numbers. Mechanistically, impaired CXCR4 desensitization disrupts cell cycle progression and osteogenic commitment of skeletal stromal/stem cells, while increasing their pro-osteoclastogenic capacities. Impaired osteogenic differentiation is evidenced in primary bone marrow stromal cells from WHIM patients. In mice, chronic treatment with the CXCR4 antagonist AMD3100 normalizes in vitro osteogenic fate of mutant skeletal stromal/stem cells and reverses in vivo the loss of skeletal cells, demonstrating that proper CXCR4 desensitization is required for the osteogenic specification of skeletal stromal/stem cells. Our study provides mechanistic insights into how CXCR4 signaling regulates the osteogenic fate of skeletal cells and the balance between bone formation and resorption.


Subject(s)
Immunologic Deficiency Syndromes , Osteoporosis , Primary Immunodeficiency Diseases , Receptors, CXCR4 , Animals , Mice , Immunologic Deficiency Syndromes/genetics , Mutation , Osteogenesis/genetics , Osteoporosis/genetics , Primary Immunodeficiency Diseases/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Humans
10.
Nat Commun ; 14(1): 588, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737440

ABSTRACT

Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders, representing high risk of progression to acute myeloid leukaemia, and frequently associated to somatic mutations, notably in the epigenetic regulator TET2. Natural Killer (NK) cells play a role in the anti-leukemic immune response via their cytolytic activity. Here we show that patients with MDS clones harbouring mutations in the TET2 gene are characterised by phenotypic defects in their circulating NK cells. Remarkably, NK cells and MDS clones from the same patient share the TET2 genotype, and the NK cells are characterised by increased methylation of genomic DNA and reduced expression of Killer Immunoglobulin-like receptors (KIR), perforin, and TNF-α. In vitro inhibition of TET2 in NK cells of healthy donors reduces their cytotoxicity, supporting its critical role in NK cell function. Conversely, NK cells from patients treated with azacytidine (#NCT02985190; https://clinicaltrials.gov/ ) show increased KIR and cytolytic protein expression, and IFN-γ production. Altogether, our findings show that, in addition to their oncogenic consequences in the myeloid cell subsets, TET2 mutations contribute to repressing NK-cell function in MDS patients.


Subject(s)
Dioxygenases , Myelodysplastic Syndromes , Humans , Methylation , Myelodysplastic Syndromes/metabolism , Killer Cells, Natural , Azacitidine/pharmacology , Receptors, KIR/genetics , Mutation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism
11.
Med Sci (Paris) ; 39(1): 23-30, 2023 Jan.
Article in French | MEDLINE | ID: mdl-36692314

ABSTRACT

CXCR4 is a chemokine receptor that plays a central role in cell migration but also in other essential processes such as the development of the immune system. Together with its ligand, the chemokine CXCL12, this signalling axis plays an important role in B lymphocyte biology from their early differentiation in the bone marrow to their activation and differentiation into antibody secreting cells, also called plasma cells. Gain-of-function mutations of CXCR4 are found in a rare immunodeficiency, the WHIM Syndrome. These mutations affect the desensitization of the receptor and lead to a gain of function in response to CXCL12. This review summarizes the role of CXCR4 in the humoral immune responses and using the WHIM Syndrome as a paradigm, highlights the critical regulatory role of CXCR4 desensitization in these processes. Indeed, recent works report that fine-tuning of CXCR4 signalling is essential to limit the extra-follicular immune response and support long term antibody-mediated protection.


Title: La signalisation de CXCR4, un rhéostat de la réponse immunitaire à médiation humorale. Abstract: CXCR4 est un récepteur de chimiokine qui joue un rôle central dans la migration cellulaire mais également dans d'autres mécanismes essentiels, tels que le développement du système immunitaire. De concert avec son ligand naturel, la chimiokine CXCL12, cet axe de signalisation joue un rôle important dans la biologie des lymphocytes B, des stades précoces de différenciation dans la moelle osseuse à leur activation et différenciation en cellules sécrétrices d'anticorps, aussi appelées plasmocytes. Des mutations gain de fonction de CXCR4 sont retrouvées dans une immunodéficience rare, le Syndrome WHIM. Ces mutations affectent le mécanisme de désensibilisation du récepteur et entraînent un gain de fonction en réponse à CXCL12. Cette revue résume le rôle de CXCR4 dans la réponse immune humorale et, à travers l'étude du Syndrome WHIM, souligne le rôle régulateur essentiel de la désensibilisation de CXCR4 dans ces processus. Des travaux récents rapportent en effet qu'une signalisation correcte de CXCR4 est essentielle pour limiter la réponse immune dite « extra-folliculaire ¼ et pour permettre une protection au long terme assurée par les anticorps.


Subject(s)
Immunologic Deficiency Syndromes , Primary Immunodeficiency Diseases , Warts , Humans , Primary Immunodeficiency Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Warts/genetics , Signal Transduction/genetics , Chemokine CXCL12/genetics , Receptors, CXCR4/genetics
12.
Proc Natl Acad Sci U S A ; 120(2): e2213056120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36595686

ABSTRACT

Despite the essential role of plasma cells in health and disease, the cellular mechanisms controlling their survival and secretory capacity are still poorly understood. Here, we identified the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) Sec22b as a unique and critical regulator of plasma cell maintenance and function. In the absence of Sec22b, plasma cells were hardly detectable and serum antibody titers were dramatically reduced. Accordingly, Sec22b-deficient mice fail to mount a protective immune response. At the mechanistic level, we demonstrated that Sec22b contributes to efficient antibody secretion and is a central regulator of plasma cell maintenance through the regulation of their transcriptional identity and of the morphology of the endoplasmic reticulum and mitochondria. Altogether, our results unveil an essential and nonredundant role for Sec22b as a regulator of plasma cell fitness and of the humoral immune response.


Subject(s)
Plasma Cells , SNARE Proteins , Mice , Animals , Plasma Cells/metabolism , R-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Endoplasmic Reticulum/metabolism , Biological Transport
13.
Immunol Lett ; 254: 6-12, 2023 02.
Article in English | MEDLINE | ID: mdl-36646289

ABSTRACT

Plasma cells and the antibodies they secrete are paramount for protection against infection but can also be implicated in diseases including autoantibody-mediated disease and multiple myeloma. Plasma cell terminal differentiation relies on a transcriptional switch and on important morphological changes. The cellular and molecular mechanisms underlying these processes are partly understood and how plasma cells manage to survive for long periods of time while secreting large quantities of antibodies remains unclear. In this review we aim to put in perspective what is known about plasma cell cellular biology to highlight the challenges faced by this field of research but also to illustrate how new opportunities may arise from the study of the fundamental mechanisms sustaining plasma cell survival and function.


Subject(s)
Multiple Myeloma , Plasma Cells , Humans , Antibodies , Cell Survival , Unfolded Protein Response
15.
Cell Rep ; 38(2): 110223, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021072

ABSTRACT

MEK1 and MEK2, the only known activators of ERK, are attractive therapeutic candidates for both cancer and autoimmune diseases. However, how MEK signaling finely regulates immune cell activation is only partially understood. To address this question, we specifically delete Mek1 in hematopoietic cells in the Mek2 null background. Characterization of an allelic series of Mek mutants reveals the presence of distinct degrees of spontaneous B cell activation, which are inversely proportional to the levels of MEK proteins and ERK activation. While Mek1 and Mek2 null mutants have a normal lifespan, 1Mek1 and 1Mek2 mutants retaining only one functional Mek1 or Mek2 allele in hematopoietic cell lineages die from glomerulonephritis and lymphoproliferative disorders, respectively. This establishes that the fine-tuning of the ERK/MAPK pathway is critical to regulate B and T cell activation and function and that each MEK isoform plays distinct roles during lymphocyte activation and disease development.


Subject(s)
Lymphocyte Activation/physiology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Alleles , Animals , B-Lymphocytes/metabolism , Female , Humans , Lymphocyte Activation/genetics , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, 129 Strain , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation , Signal Transduction/physiology , T-Lymphocytes/metabolism
16.
Eur J Immunol ; 52(1): 10-23, 2022 01.
Article in English | MEDLINE | ID: mdl-34694625

ABSTRACT

Long considered a homogeneous population dedicated to antibody secretion, plasma cell phenotypic and functional heterogeneity is increasingly recognized. Plasma cells were first segregated based on their maturation level, but the complexity of this subset might well be underestimated by this simple dichotomy. Indeed, in the last decade new functions have been attributed to plasma cells including but not limited to cytokine secretion. However, a proper characterization of plasma cell heterogeneity has remained elusive partly due to technical issues and cellular features that are specific to this cell type. Cell intrinsic and cell extrinsic signals could be at the origin of this heterogeneity. Recent advances in technologies such as single cell RNA-seq, ATAC-seq, or ChIP-seq on low cell numbers helped to elucidate the fate decision in other cell lineages and similar approaches could be implemented to evaluate the heterogeneous fate of activated B cells in health and disease. Here, we summarized published work shedding some lights on the stimuli and genetic program shaping B-cell terminal differentiation at the single cell level in mice and men. We also discuss the fate and heterogeneity of plasma cells during immune responses, vaccination, and in the frame of human plasma cell disorders.


Subject(s)
Cell Differentiation , Chromatin Immunoprecipitation Sequencing , Immune System Diseases , Plasma Cells/immunology , RNA-Seq , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology
18.
Leukemia ; 35(10): 2895-2905, 2021 10.
Article in English | MEDLINE | ID: mdl-34363012

ABSTRACT

Aberrant CXCR4 activity has been implicated in lymphoma pathogenesis, disease progression, and resistance to therapies. Using a mouse model with a gain-of-function CXCR4 mutation (CXCR4C1013G) that hyperactivates CXCR4 signaling, we identified CXCR4 as a crucial activator of multiple key oncogenic pathways. CXCR4 hyperactivation resulted in an expansion of transitional B1 lymphocytes, which represent the precursors of chronic lymphocytic leukemia (CLL). Indeed, CXCR4 hyperactivation led to a significant acceleration of disease onset and a more aggressive phenotype in the murine Eµ-TCL1 CLL model. Hyperactivated CXCR4 signaling cooperated with TCL1 to cause a distinct oncogenic transcriptional program in B cells, characterized by PLK1/FOXM1-associated pathways. In accordance, Eµ-TCL1;CXCR4C1013G B cells enriched a transcriptional signature from patients with Richter's syndrome, an aggressive transformation of CLL. Notably, MYC activation in aggressive lymphoma was associated with increased CXCR4 expression. In line with this finding, additional hyperactive CXCR4 signaling in the Eµ-Myc mouse, a model of aggressive B-cell cancer, did not impact survival. In summary, we here identify CXCR4 hyperactivation as a co-driver of an aggressive lymphoma phenotype.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mutation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Receptors, CXCR4/metabolism , Animals , Cell Cycle Proteins/genetics , Disease Progression , Female , Forkhead Box Protein M1/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptors, CXCR4/genetics , Polo-Like Kinase 1
19.
Front Cell Dev Biol ; 9: 639022, 2021.
Article in English | MEDLINE | ID: mdl-34386488

ABSTRACT

Several studies have established the crucial role of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase pathway in hematopoietic cell proliferation and differentiation. MEK1 and MEK2 phosphorylate and activate ERK1 and ERK2. However, whether MEK1 and MEK2 differentially regulate these processes is unknown. To define the function of Mek genes in the activation of the ERK pathway during hematopoiesis, we generated a mutant mouse line carrying a hematopoietic-specific deletion of the Mek1 gene function in a Mek2 null background. Inactivation of both Mek1 and Mek2 genes resulted in death shortly after birth with a severe anemia revealing the essential role of the ERK pathway in erythropoiesis. Mek1 and Mek2 functional ablation also affected lymphopoiesis and myelopoiesis. In contrast, mice that retained one functional Mek1 (1Mek1) or Mek2 (1Mek2) allele in hematopoietic cells were viable and fertile. 1Mek1 and 1Mek2 mutants showed mild signs of anemia and splenomegaly, but the half-life of their red blood cells and the response to erythropoietic stress were not altered, suggesting a certain level of Mek redundancy for sustaining functional erythropoiesis. However, subtle differences in multipotent progenitor distribution in the bone marrow were observed in 1Mek1 mice, suggesting that the two Mek genes might differentially regulate early hematopoiesis.

20.
Front Immunol ; 12: 658535, 2021.
Article in English | MEDLINE | ID: mdl-33936091

ABSTRACT

The bone marrow is a complex ecosystem in which hematopoietic and non-hematopoietic cells reside. In this review, we discuss the bone marrow niches in mice that facilitate the survival, maintenance, and differentiation of cells of hematopoietic origin based on the recent literature. Our review places a special focus on the hematopoietic multipotent progenitors and on plasma cells, corresponding to the last stage of the B-cell lineage, that play a key role in the humoral memory response. We highlight the similarities between the microenvironments necessary for the establishment and the maintenance of these two immune cell subsets, and how the chemokine CXCL12/CXCR4 signaling axis contributes to these processes. Finally, we bring elements to address the following question: are multipotent progenitors and plasma cells neighbors or roommates within the bone marrow?


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Lymphopoiesis , Plasma Cells/cytology , Plasma Cells/metabolism , Animals , Biomarkers , Bone Marrow/blood supply , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Communication , Cellular Microenvironment , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...