Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Life Sci Alliance ; 6(4)2023 04.
Article de Anglais | MEDLINE | ID: mdl-36717247

RÉSUMÉ

Systemic inflammation halts lymphopoiesis and prioritizes myeloid cell production. How blood cell production switches from homeostasis to emergency myelopoiesis is incompletely understood. Here, we show that lymphotoxin-ß receptor (LTßR) signaling in combination with TNF and IL-1 receptor signaling in bone marrow mesenchymal stem cells (MSCs) down-regulates Il7 expression to shut down lymphopoiesis during systemic inflammation. LTßR signaling in MSCs also promoted CCL2 production during systemic inflammation. Pharmacological or genetic blocking of LTßR signaling in MSCs partially enabled lymphopoiesis and reduced monocyte numbers in the spleen during systemic inflammation, which correlated with reduced survival during systemic bacterial and viral infections. Interestingly, lymphotoxin-α1ß2 delivered by B-lineage cells, and specifically by mature B cells, contributed to promote Il7 down-regulation and reduce MSC lymphopoietic activity. Our studies revealed an unexpected role of LTßR signaling in MSCs and identified recirculating mature B cells as an important regulator of emergency myelopoiesis.


Sujet(s)
Cellules souches mésenchymateuses , Myélopoïèse , Humains , Interleukine-7 , Lymphocytes B/métabolisme , Cellules souches mésenchymateuses/métabolisme , Inflammation/métabolisme
2.
ZDM ; 55(1): 109-118, 2023.
Article de Anglais | MEDLINE | ID: mdl-36532825

RÉSUMÉ

The mathematical medium of data visualization and other data representations (DV) has served as a primary means of communicating about the COVID-19 crisis. DVs about the pandemic are highly visible across news journalism and include an increasingly innovative and diverse set of representational forms. These representational forms employ multimodal, interactive, and narrative elements, among others, that create new possibilities for data storytelling. Building on current efforts to expand the teaching and learning of data practices in K-12 mathematics education, we argue that innovative DVs create new opportunities for teaching and learning mathematics, particularly during times of crisis. We illustrate our argument using three examples of innovative DVs from news journalism. We discuss how these DVs could serve as complementary resources alongside conventional graphs to support students as they use mathematics and mathematical representations to make sense of crises such as the COVID-19 pandemic. Our commentary seeks to bring current trends in data representation to bear in mathematics education. Leveraging such trends offers artifacts useful for teaching and opens up space for elevating emotion and experience as important aspects of mathematics curricula.

3.
Sci Immunol ; 7(75): eabo3170, 2022 09 23.
Article de Anglais | MEDLINE | ID: mdl-36149943

RÉSUMÉ

Gain-of-function (GOF) mutations in CXCR4 cause WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome, characterized by infections, leukocyte retention in bone marrow (BM), and blood leukopenias. B lymphopenia is evident at early progenitor stages, yet why do CXCR4 GOF mutations that cause B (and T) lymphopenia remain obscure? Using a CXCR4 R334X GOF mouse model of WHIM syndrome, we showed that lymphopoiesis is reduced because of a dysregulated mesenchymal stem cell (MSC) transcriptome characterized by a switch from an adipogenic to an osteolineage-prone program with limited lymphopoietic activity. We identify lymphotoxin beta receptor (LTßR) as a critical pathway promoting interleukin-7 (IL-7) down-regulation in MSCs. Blocking LTßR or CXCR4 signaling restored IL-7 production and B cell development in WHIM mice. LTßR blocking also increased production of IL-7 and B cell activating factor (BAFF) in secondary lymphoid organs (SLOs), increasing B and T cell numbers in the periphery. These studies revealed that LTßR signaling in BM MSCs and SLO stromal cells limits the lymphocyte compartment size.


Sujet(s)
Déficits immunitaires , Lymphopénie , Animaux , Facteur d'activation des lymphocytes B , Déficits immunitaires/complications , Déficits immunitaires/génétique , Interleukine-7 , Récepteur à la lymphotoxine-bêta , Souris , Maladies d'immunodéficience primaire , Niche de cellules souches , Lymphocytes T , Verrues
4.
Front Immunol ; 11: 600127, 2020.
Article de Anglais | MEDLINE | ID: mdl-33324418

RÉSUMÉ

Studies over the last couple of decades have shown that hematopoietic stem cells (HSCs) are critically dependent on cytokines such as Stem Cell Factor and other signals provided by bone marrow niches comprising of mesenchymal stem and progenitor cells (MSPCs) and endothelial cells (ECs). Because of their critical roles in HSC maintenance the niches formed by MSPCs and ECs are commonly referred to as HSC niches. For the most part, the signals required for HSC maintenance act in a short-range manner, which imposes the necessity for directional and positional cues in order for HSCs to localize and be retained properly in stem cell niches. The chemokine CXCL12 and its Gαi protein coupled receptor CXCR4, besides promoting HSC quiescence directly, also play instrumental roles in enabling HSCs to access bone marrow stem cell niches. Recent studies have revealed, however, that HSC niches also provide a constellation of hematopoietic cytokines that are critical for the production of most, if not all, blood cell types. Some hematopoietic cytokines, namely IL-7 and IL-15 produced by HSC niches, are not only required for lymphopoiesis but are also essential for memory T cell maintenance. Consequently, hematopoietic progenitors and differentiated immune cells, such as memory T cell subsets, also depend on the CXCL12/CXCR4 axis for migration into bone marrow and interactions with MSPCs and ECs. Similarly, subsets of antibody-secreting plasma cells also reside in close association with CXCL12-producing MSPCs in the bone marrow and require the CXCR4/CXCL12 axis for survival and long-term maintenance. Collectively, these studies demonstrate a broad range of key physiological roles, spanning blood cell production and maintenance of immunological memory, that are orchestrated by stem cell niches through a common and simple mechanism: CXCL12/CXCR4-mediated cell recruitment followed by receipt of a maintenance and/or instructive signal. A fundamental flaw of this type of cellular organization is revealed by myeloid and lymphoid leukemias, which target stem cell niches and induce profound transcriptomic changes that result in reduced hematopoietic activity and altered mesenchymal cell differentiation.


Sujet(s)
Cellules souches hématopoïétiques/immunologie , Mémoire immunologique , Transduction du signal/immunologie , Niche de cellules souches/immunologie , Animaux , Cellules endothéliales/immunologie , Humains , Cellules souches mésenchymateuses/immunologie
5.
Adv Immunol ; 134: 47-88, 2017.
Article de Anglais | MEDLINE | ID: mdl-28413023

RÉSUMÉ

B lymphocytes develop from hematopoietic stem cells (HSCs) in specialized bone marrow niches composed of rare mesenchymal lineage stem/progenitor cells (MSPCs) and sinusoidal endothelial cells. These niches are defined by function and location: MSPCs are mostly perisinusoidal cells that together with a small subset of sinusoidal endothelial cells express stem cell factor, interleukin-7 (IL-7), IL-15, and the highest amounts of CXCL12 in bone marrow. Though rare, MSPCs are morphologically heterogeneous, highly reticular, and form a vast cellular network in the bone marrow parenchyma capable of interacting with large numbers of hematopoietic cells. HSCs, downstream multipotent progenitor cells, and common lymphoid progenitor cells utilize CXCR4 to fine-tune access to critical short-range growth factors provided by MSPCs for their long-term maintenance and/or multilineage differentiation. In later stages, developing B lymphocytes use CXCR4 to navigate the bone marrow parenchyma, and predominantly cannabinoid receptor-2 for positioning within bone marrow sinusoids, prior to being released into peripheral blood circulation. In the final stages of differentiation, transitional B cells migrate to the spleen where they preferentially undergo further rounds of differentiation until selection into the mature B cell pool occurs. This bottleneck purges up to 97% of all developing B cells in a peripheral selection process that is heavily controlled not only by the intensity of BCR signaling and access to BAFF but also by the proper functioning of the B cell motility machinery.


Sujet(s)
Lymphocytes B/cytologie , Différenciation cellulaire/immunologie , Chimiokines/immunologie , Cellules souches hématopoïétiques/cytologie , Lymphopoïèse/immunologie , Animaux , Lymphocytes B/immunologie , Cellules souches hématopoïétiques/immunologie , Humains
6.
Immunity ; 45(6): 1219-1231, 2016 12 20.
Article de Anglais | MEDLINE | ID: mdl-27913094

RÉSUMÉ

Hematopoietic stem cells (HSCs) self-renew in bone marrow niches formed by mesenchymal progenitors and endothelial cells expressing the chemokine CXCL12, but whether a separate niche instructs multipotent progenitor (MPP) differentiation remains unclear. We show that MPPs resided in HSC niches, where they encountered lineage-instructive differentiation signals. Conditional deletion of the chemokine receptor CXCR4 in MPPs reduced differentiation into common lymphoid progenitors (CLPs), which decreased lymphopoiesis. CXCR4 was required for CLP positioning near Interleukin-7+ (IL-7) cells and for optimal IL-7 receptor signaling. IL-7+ cells expressed CXCL12 and the cytokine SCF, were mesenchymal progenitors capable of differentiation into osteoblasts and adipocytes, and comprised a minor subset of sinusoidal endothelial cells. Conditional Il7 deletion in mesenchymal progenitors reduced B-lineage committed CLPs, while conditional Cxcl12 or Scf deletion from IL-7+ cells reduced HSC and MPP numbers. Thus, HSC maintenance and multilineage differentiation are distinct cell lineage decisions that are both controlled by HSC niches.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules souches hématopoïétiques/cytologie , Cellules souches multipotentes/cytologie , Niche de cellules souches/physiologie , Animaux , Lignage cellulaire/physiologie , Séparation cellulaire , Chimiokine CXCL2/métabolisme , Cytométrie en flux , Interleukine-7/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques
7.
Tissue Eng Part A ; 20(1-2): 424-33, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24020641

RÉSUMÉ

Stem cells have tremendous potential for treating various human diseases. Protocols have been established to differentiate stem cells into specific lineages through the provision of signals in the form of growth factors, cytokines, or small molecules. Herein we investigate an alternative strategy for directed differentiation of human embryonic stem cells (hESCs)--extracellular-matrix (ECM) mediated differentiation. Decellularized ECM and conditioned media from the appropriate committed cell lines are used to differentiate stem cells to the required phenotype. Applying this strategy to differentiate hESCs to pancreatic beta cells, we have obtained functional cells that secreted insulin in a glucose-responsive manner, and were able to recover normoglycemia in a streptozotocin (STZ)-induced diabetic mouse model. ECM-mediated differentiation was also demonstrated to be effective for the differentiation of hESCs into kidney tubule cells and cardiomyocytes. Gene expression studies suggested the involvement of integrins and catenins in the beta cell differentiation process; in particular, α1, αv, and ß1 integrins, and ß-catenin showed the highest upregulation. To further elucidate the biochemical and mechanical cues that have led to effective hESC differentiation to beta cells, we have employed an artificial system that allowed for variation of matrix stiffness and combination of individual ECM proteins at various ratios. The differentiation response of hESCs to the native ECM could be approximated by optimizing this system.


Sujet(s)
Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Matrice extracellulaire/métabolisme , Cellules à insuline/cytologie , Animaux , Lignée cellulaire , Diabète expérimental/anatomopathologie , Cellules souches embryonnaires/métabolisme , Protéines de la matrice extracellulaire/métabolisme , Régulation de l'expression des gènes , Humains , Cellules à insuline/métabolisme , Intégrines/métabolisme , Souris , Souris SCID , Sous-unités de protéines/métabolisme , Rats , Transduction du signal , bêta-Caténine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE