Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Immunol ; 204(10): 2685-2696, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32229537

ABSTRACT

Grasp55 is a ubiquitous Golgi stacking protein involved in autophagy, protein trafficking, and glucose deprivation sensing. The function of Grasp55 in protein trafficking has been attributed to its PDZ-mediated interaction with the C-terminal PDZ-binding motifs of protein cargos. We have recently shown that such an interaction occurs between Grasp55 and the adhesion molecule Jam-C, which plays a central role in stemness maintenance of hematopoietic and spermatogenic cells. Accordingly, we have found that Grasp55-deficient mice suffer from spermatogenesis defects similar to Jam-C knockout mice. However, whether Grasp55 is involved in the maintenance of immunohematopoietic homeostasis through regulation of protein transport and Jam-C expression remains unknown. In this study, we show that Grasp55 deficiency does not affect hematopoietic stem cell differentiation, engraftment, or mobilization, which are known to depend on expression of Grasp55-dependent protein cargos. In contrast, using an Myc-dependent leukemic model addicted to autophagy, we show that knockdown of Grasp55 in leukemic cells reduces spleen and bone marrow tumor burden upon i.v. leukemic engraftment. This is not due to reduced homing of Grasp55-deficient cells to these organs but to increased spontaneous apoptosis of Grasp55-deficient leukemic cells correlated with increased sensitivity of the cells to glucose deprivation. These results show that Grasp55 plays a role in Myc-transformed hematopoietic cells but not in normal hematopoietic cells in vivo.


Subject(s)
Golgi Apparatus/pathology , Golgi Matrix Proteins/metabolism , Leukemia/metabolism , Animals , Apoptosis/genetics , Autophagy , Carcinogenesis , Cell Survival , Golgi Matrix Proteins/genetics , Hematopoiesis/genetics , Leukemia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport , Proto-Oncogene Proteins c-myc/metabolism , Tumor Burden
2.
Proc Natl Acad Sci U S A ; 116(1): 211-216, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30559202

ABSTRACT

Bone marrow (BM) produces all blood and immune cells deriving from hematopoietic stem cells (HSCs). The decrease of immune cell production during aging is one of the features of immunosenescence. The impact of redox dysregulation in BM aging is still poorly understood. Here we use TP53INP1-deficient (KO) mice endowed with chronic oxidative stress to assess the influence of aging-associated redox alterations in BM homeostasis. We show that TP53INP1 deletion has no impact on aging-related accumulation of HSCs. In contrast, the aging-related contraction of the lymphoid compartment is mitigated in TP53INP1 KO mice. B cells that accumulate in old KO BM are differentiating cells that can mature into functional B cells. Importantly, this phenotype results from B cell-intrinsic events associated with defective redox control. Finally, we show that oxidative stress in aged TP53INP1-deficient mice maintains STAT5 expression and activation in early B cells, driving high Pax5 expression, which provides a molecular mechanism for maintenance of B cell development upon aging.


Subject(s)
B-Lymphocytes/physiology , Bone Marrow/physiology , Lymphopoiesis , Nuclear Proteins/deficiency , Receptors, Interleukin-7/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Aging/physiology , Animals , B-Lymphocytes/metabolism , Bone Marrow/metabolism , Lymphopoiesis/physiology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress
3.
Proc Natl Acad Sci U S A ; 115(41): 10357-10362, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30257940

ABSTRACT

PAX5 is a well-known haploinsufficient tumor suppressor gene in human B-cell precursor acute lymphoblastic leukemia (B-ALL) and is involved in various chromosomal translocations that fuse a part of PAX5 with other partners. However, the role of PAX5 fusion proteins in B-ALL initiation and transformation is ill-known. We previously reported a new recurrent t(7;9)(q11;p13) chromosomal translocation in human B-ALL that juxtaposed PAX5 to the coding sequence of elastin (ELN). To study the function of the resulting PAX5-ELN fusion protein in B-ALL development, we generated a knockin mouse model in which the PAX5-ELN transgene is expressed specifically in B cells. PAX5-ELN-expressing mice efficiently developed B-ALL with an incidence of 80%. Leukemic transformation was associated with recurrent secondary mutations on Ptpn11, Kras, Pax5, and Jak3 genes affecting key signaling pathways required for cell proliferation. Our functional studies demonstrate that PAX5-ELN affected B-cell development in vitro and in vivo featuring an aberrant expansion of the pro-B cell compartment at the preleukemic stage. Finally, our molecular and computational approaches identified PAX5-ELN-regulated gene candidates that establish the molecular bases of the preleukemic state to drive B-ALL initiation. Hence, our study provides a new in vivo model of human B-ALL and strongly implicates PAX5 fusion proteins as potent oncoproteins in leukemia development.


Subject(s)
Elastin/genetics , Oncogene Proteins, Fusion/genetics , PAX5 Transcription Factor/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Elastin/metabolism , Gene Expression Regulation, Leukemic , Gene Knock-In Techniques , Janus Kinase 3/genetics , Mice, Transgenic , Mutation , Neoplasms, Experimental , Oncogene Proteins, Fusion/metabolism , PAX5 Transcription Factor/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins p21(ras)/genetics
4.
Int J Mol Sci ; 19(8)2018 Aug 10.
Article in English | MEDLINE | ID: mdl-30103411

ABSTRACT

After birth, the development of hematopoietic cells occurs in the bone marrow. Hematopoietic differentiation is finely tuned by cell-intrinsic mechanisms and lineage-specific transcription factors. However, it is now clear that the bone marrow microenvironment plays an essential role in the maintenance of hematopoietic stem cells (HSC) and their differentiation into more mature lineages. Mesenchymal and endothelial cells contribute to a protective microenvironment called hematopoietic niches that secrete specific factors and establish a direct contact with developing hematopoietic cells. A number of recent studies have addressed in mouse models the specific molecular events that are involved in the cellular crosstalk between hematopoietic subsets and their niches. This has led to the concept that hematopoietic differentiation and commitment towards a given hematopoietic pathway is a dynamic process controlled at least partially by the bone marrow microenvironment. In this review, we discuss the evolving view of murine hematopoietic⁻stromal cell crosstalk that is involved in HSC maintenance and commitment towards B cell differentiation.


Subject(s)
Bone Marrow/metabolism , Cell Differentiation/physiology , Lymphopoiesis/physiology , Precursor Cells, B-Lymphoid/metabolism , Stem Cell Niche/physiology , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Precursor Cells, B-Lymphoid/cytology , Transcription Factors/metabolism
5.
Cell Mol Life Sci ; 73(4): 687-703, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26495446

ABSTRACT

Hematopoiesis takes place in the bone marrow of adult mammals and is the process by which blood cells are replenished every day throughout life. Differentiation of hematopoietic cells occurs in a stepwise manner through intermediates of differentiation that could be phenotypically identified. This has allowed establishing hematopoietic cell classification with hematopoietic stem cells (HSCs) at the top of the hierarchy. HSCs are mostly quiescent and serve as a reservoir for maintenance of lifelong hematopoiesis. Over recent years, it has become increasingly clear that HSC quiescence is not only due to intrinsic properties, but is also mediated by cognate interactions between HSCs and surrounding cells within micro-anatomical sites called "niches". This hematopoietic/stromal crosstalk model also applies to more mature progenitors such as B cell progenitors, which are thought to reside in distinct "niches". This prompted many research teams to search for specific molecular mechanisms supporting leuko-stromal crosstalk in the bone marrow and acting at specific stage of differentiation to regulate hematopoietic homeostasis. Here, we review recent data on adhesion mechanisms involved in HSCs and B cell progenitors interactions with surrounding bone marrow stromal cells.


Subject(s)
B-Lymphocytes/cytology , Cell Communication , Hematopoietic Stem Cells/cytology , Stem Cell Niche , Animals , B-Lymphocytes/metabolism , Cadherins/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Integrins/metabolism , Mucins/metabolism , Selectins/metabolism
6.
J Immunol ; 190(11): 5526-33, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23616571

ABSTRACT

Galectin-1 (GAL1) is an S-type lectin with multiple functions, including the control of B cell homeostasis. GAL1 expression was reported to be under the control of the plasma cell master regulator BLIMP-1. GAL1 was detected at the protein level in LPS-stimulated B cells and was shown to promote Ig secretion in vitro. However, the pattern of GAL1 expression and function of GAL1 in B cells in vivo are still unclear. In this study, we show that, among B cells, GAL1 is only expressed by differentiating plasma cells following T-dependent or T-independent immunization. Using GAL1-deficient mice we demonstrate that GAL1 expression is required for the maintenance of Ag-specific Ig titers and Ab-secreting cell numbers. Using an in vitro differentiation assay we find that GAL1-deficient plasmablasts can develop normally but die rapidly, through caspase 8 activation, under serum starvation-induced death conditions. TUNEL assays show that in vivo-generated GAL1-deficient plasma cells exhibit an increased sensitivity to apoptosis. Taken together, our data indicate that endogenous GAL1 supports plasma cell survival and participates in the regulation of the humoral immune response.


Subject(s)
Galectin 1/metabolism , Homeostasis/immunology , Immunity, Humoral , Plasma Cells/immunology , Plasma Cells/metabolism , Animals , Antigens/immunology , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Galectin 1/genetics , Homeostasis/genetics , Immunity, Humoral/genetics , Immunoglobulin G/blood , Immunophenotyping , Lymphocyte Count , Mice , Mice, Knockout , Plasma Cells/cytology , Spleen/immunology , Spleen/metabolism
7.
Cell Rep ; 43(8): 114541, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39058594

ABSTRACT

Galectins are glycan-binding proteins translating the sugar-encoded information of cellular glycoconjugates into physiological activities, including immunity, cell migration, and signaling. Galectins also interact with non-glycosylated partners in the extracellular milieu, among which the pre-B cell receptor (pre-BCR) during B cell development. How these interactions might interplay with the glycan-decoding function of galectins is unknown. Here, we perform NMR experiments on native membranes to monitor Gal-1 binding to physiological cell surface ligands. We show that pre-BCR interaction changes Gal-1 binding to glycosylated pre-B cell surface receptors. At the molecular and cellular levels, we identify α2,3-sialylated motifs as key targeted epitopes. This targeting occurs through a selectivity switch increasing Gal-1 contacts with α2,3-sialylated poly-N-acetyllactosamine upon pre-BCR interaction. Importantly, we observe that this switch is involved in the regulation of pre-BCR activation. Altogether, this study demonstrates that interactions to non-glycosylated proteins regulate the glycan-decoding functions of galectins at the cell surface.

8.
J Exp Med ; 221(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37930337

ABSTRACT

B cell acute lymphoblastic leukemia (B-ALL) is a multistep disease characterized by the hierarchical acquisition of genetic alterations. However, the question of how a primary oncogene reprograms stem cell-like properties in committed B cells and leads to a preneoplastic population remains unclear. Here, we used the PAX5::ELN oncogenic model to demonstrate a causal link between the differentiation blockade, the self-renewal, and the emergence of preleukemic stem cells (pre-LSCs). We show that PAX5::ELN disrupts the differentiation of preleukemic cells by enforcing the IL7r/JAK-STAT pathway. This disruption is associated with the induction of rare and quiescent pre-LSCs that sustain the leukemia-initiating activity, as assessed using the H2B-GFP model. Integration of transcriptomic and chromatin accessibility data reveals that those quiescent pre-LSCs lose B cell identity and reactivate an immature molecular program, reminiscent of human B-ALL chemo-resistant cells. Finally, our transcriptional regulatory network reveals the transcription factor EGR1 as a strong candidate to control quiescence/resistance of PAX5::ELN pre-LSCs as well as of blasts from human B-ALL.


Subject(s)
Burkitt Lymphoma , Leukemia , Humans , Janus Kinases , STAT Transcription Factors , Signal Transduction , Stem Cells
9.
Blood ; 117(24): 6552-61, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21511956

ABSTRACT

In the bone marrow (BM), stromal cells constitute a supportive tissue indispensable for the generation of pro-B/pre-BI, pre-BII, and immature B lymphocytes. IL-7-producing stromal cells constitute a cellular niche for pro-B/pre-BI cells, but no specific stromal cell microenvironment was identified for pre-BII cells expressing a functional pre-B cell receptor (pre-BCR). However expression of the pre-BCR represents a crucial checkpoint during B-cell development. We recently demonstrated that the stromal cell derived-galectin1 (GAL1) is a ligand for the pre-BCR, involved in the proliferation and differentiation of normal mouse pre-BII cells. Here we show that nonhematopoietic osteoblasts and reticular cells in the BM express GAL1. We observed that pre-BII cells, unlike the other B-cell subsets, were specifically localized in close contact with GAL1(+) reticular cells. We also determined that IL-7(+) and GAL1(+) cells represent 2 distinct mesenchymal populations with different BM localization. These results demonstrate the existence of a pre-BII specific stromal cell niche and indicate that early B cells move from IL-7(+) to GAL1(+) supportive BM niches during their development.


Subject(s)
Bone Marrow , Galectin 1/metabolism , Precursor Cells, B-Lymphoid/physiology , Stem Cell Niche/physiology , Stromal Cells/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Differentiation/immunology , Cells, Cultured , Green Fluorescent Proteins/genetics , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pre-B Cell Receptors/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Stem Cell Niche/cytology , Stem Cell Niche/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
10.
Blood ; 118(17): 4609-19, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21868569

ABSTRACT

In adult mammals, hematopoietic stem cells (HSCs) reside in the bone marrow (BM) and are maintained in a quiescent and undifferentiated state through adhesive interactions with specialized microenvironmental niches. Although junctional adhesion molecule-C (JAM-C) is expressed by HSCs, its function in adult hematopoiesis remains elusive. Here, we show that HSCs adhere to JAM-B expressed by BM stromal cells in a JAM-C dependent manner. The interaction regulates the interplay between HSCs and BM stromal cells as illustrated by the decreased pool of quiescent HSCs observed in jam-b deficient mice. We further show that this is probably because of alterations of BM stromal compartments and changes in SDF-1α BM content in jam-b(-/-) mice, suggesting that JAM-B is an active player in the maintenance of the BM stromal microenvironment.


Subject(s)
Bone Marrow Cells/physiology , Cell Adhesion Molecules/physiology , Hematopoietic Stem Cells/physiology , Immunoglobulins/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
J Immunol ; 187(4): 1835-44, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21746964

ABSTRACT

B cells are subjected to selection at multiple checkpoints during their development. The selection of Ab H chains is difficult to study because of the large diversity of the CDR3. To study the selection of individual Ab H chain V region genes (V(H)), we performed CDR3 spectratyping of ∼ 75-300 rearrangements per individual V(H) in C57BL6/J mice. We measured the fraction of rearrangements that were in-frame in B cell DNA. We demonstrate that individual V(H)s have different fractions of in-frame rearrangements (IF fractions) ranging from 10 to 90% and that these IF fractions are reproducible in different mice. For most V(H)s, the IF fraction in pro-B cells approximated 33% and then shifted to the nearly final (mature) B cell value by the cycling pre-B cell stage. The frequency of high in-frame (IF) V(H) usage increased in cycling pre-B cells compared with that in pro-B cells, whereas this did not occur for low IF V(H)s. The IF fraction did not shift as much in BCR-expressing B cells and was minimally affected by L chain usage for most V(H). High IF clan II/III V(H)s share more positively charged CDR2 sequences, whereas high IF clan I J558 CDR2 sequences are diverse. These data indicate that individual V(H)s are subjected to differential selection, that V(H) IF fraction is mainly established through pre-BCR-mediated selection, that it may operate differently in clan I versus II/III V(H)s, and that it has a lasting influence on the Ab repertoire.


Subject(s)
Complementarity Determining Regions/metabolism , DNA/metabolism , Precursor Cells, B-Lymphoid/metabolism , Somatic Hypermutation, Immunoglobulin/physiology , Animals , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , DNA/genetics , DNA/immunology , Gene Expression Regulation/physiology , Mice , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Proto-Oncogene Proteins c-bcr/biosynthesis , Proto-Oncogene Proteins c-bcr/genetics , Proto-Oncogene Proteins c-bcr/immunology
12.
iScience ; 26(4): 106385, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37009219

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) reflects the malignant counterpart of developing B cells in the bone marrow (BM). Despite tremendous progress in B-ALL treatment, the overall survival of adults at diagnosis and patients at all ages after relapse remains poor. Galectin-1 (GAL1) expressed by BM supportive niches delivers proliferation signals to normal pre-B cells through interaction with the pre-B cell receptor (pre-BCR). Here, we asked whether GAL1 gives non-cell autonomous signals to pre-BCR+ pre-B ALL, in addition to cell-autonomous signals linked to genetic alterations. In syngeneic and patient-derived xenograft (PDX) murine models, murine and human pre-B ALL development is influenced by GAL1 produced by BM niches through pre-BCR-dependent signals, similarly to normal pre-B cells. Furthermore, targeting pre-BCR signaling together with cell-autonomous oncogenic pathways in pre-B ALL PDX improved treatment response. Our results show that non-cell autonomous signals transmitted by BM niches represent promising targets to improve B-ALL patient survival.

13.
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
14.
Blood ; 113(23): 5878-86, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19329777

ABSTRACT

Activation of the pre-B-cell receptor (pre-BCR) in the bone marrow depends on both tonic and ligand-induced signaling and leads to pre-BII-cell proliferation and differentiation. Using normal mouse bone marrow pre-BII cells, we demonstrate that the ligand-induced pre-BCR activation depends on pre-BCR/galectin-1/integrin interactions leading to pre-BCR clustering at the pre-BII/stromal cell synapse. In contrast, heparan sulfates, shown to be pre-BCR ligands in mice, are not implicated in pre-BCR relocalization. Inhibition of pre-BCR/galectin-1/integrin interactions has functional consequences, since pre-BII-cell proliferation and differentiation are impaired in an in vitro B-cell differentiation assay, without affecting cellular apoptosis. Most strikingly, although galectin-1-deficient mice do not show an apparent B-cell phenotype, the kinetics of de novo B-cell reconstitution after hydroxyurea treatment indicates a specific delay in pre-BII-cell recovery due to a decrease in pre-BII-cell differentiation and proliferation. Thus, although it remains possible that the pre-BCR interacts with other ligands, these results highlight the role played by the stromal cell-derived galectin-1 for the efficient development of normal pre-BII cells and suggest the existence of pre-BII-specific stromal cell niches in normal bone marrow.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Galectin 1/metabolism , Stromal Cells/cytology , Stromal Cells/immunology , Animals , B-Lymphocytes/metabolism , Cell Line , Cell Proliferation , Galectin 1/deficiency , Galectin 1/genetics , Integrins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pre-B Cell Receptors/immunology , Stromal Cells/metabolism
15.
Front Immunol ; 12: 784691, 2021.
Article in English | MEDLINE | ID: mdl-34956214

ABSTRACT

B-cell non-Hodgkin lymphoma (B-NHL) evolution and treatment are complicated by a high prevalence of relapses primarily due to the ability of malignant B cells to interact with tumor-supportive lymph node (LN) and bone marrow (BM) microenvironments. In particular, progressive alterations of BM stromal cells sustain the survival, proliferation, and drug resistance of tumor B cells during diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and chronic lymphocytic leukemia (CLL). The current review describes how the crosstalk between BM stromal cells and lymphoma tumor cells triggers the establishment of the tumor supportive niche. DLBCL, FL, and CLL display distinct patterns of BM involvement, but in each case tumor-infiltrating stromal cells, corresponding to cancer-associated fibroblasts, exhibit specific phenotypic and functional features promoting the recruitment, adhesion, and survival of tumor cells. Tumor cell-derived extracellular vesicles have been recently proposed as playing a central role in triggering initial induction of tumor-supportive niches, notably within the BM. Finally, the disruption of the BM stroma reprogramming emerges as a promising therapeutic option in B-cell lymphomas. Targeting the crosstalk between BM stromal cells and malignant B cells, either through the inhibition of stroma-derived B-cell growth factors or through the mobilization of clonal B cells outside their supportive BM niche, should in particular be further evaluated as a way to avoid relapses by abrogating resistance niches.


Subject(s)
B-Lymphocytes/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphoma, Follicular/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Mesenchymal Stem Cells/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow/pathology , Cell Communication/immunology , Cell Differentiation/immunology , Coculture Techniques , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Stem Cell Niche/immunology , Tumor Cells, Cultured , Tumor Microenvironment/immunology
16.
Methods Mol Biol ; 2308: 73-81, 2021.
Article in English | MEDLINE | ID: mdl-34057715

ABSTRACT

Flow cytometry has been widely used to detect a single event by means of multiparametric fluorescence measurements. Here we describe a method to analyze subsets of hematopoietic stem and progenitor cells isolated from long bones of mice. We further show that this method allows for comparing JAM-C protein expression between subsets of hematopoietic stem and progenitor cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Immunoglobulins/metabolism , Multipotent Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Lineage , Mice , Phenotype
17.
Front Immunol ; 12: 756231, 2021.
Article in English | MEDLINE | ID: mdl-34867994

ABSTRACT

In the bone marrow (BM) of adult mammals, haematopoietic stem cells (HSCs) are retained in micro-anatomical structures by adhesion molecules that regulate HSC quiescence, proliferation and commitment. During decades, researchers have used engraftment to study the function of adhesion molecules in HSC's homeostasis regulation. Since the 90's, progress in genetically engineered mouse models has allowed a better understanding of adhesion molecules involved in HSCs regulation by BM niches and raised questions about the role of adhesion mechanisms in conferring drug resistance to cancer cells nested in the BM. This has been especially studied in acute myeloid leukaemia (AML) which was the first disease in which the concept of cancer stem cell (CSC) or leukemic stem cells (LSCs) was demonstrated. In AML, it has been proposed that LSCs propagate the disease and are able to replenish the leukemic bulk after complete remission suggesting that LSC may be endowed with drug resistance properties. However, whether such properties are due to extrinsic or intrinsic molecular mechanisms, fully or partially supported by molecular crosstalk between LSCs and surrounding BM micro-environment is still matter of debate. In this review, we focus on adhesion molecules that have been involved in HSCs or LSCs anchoring to BM niches and discuss if inhibition of such mechanism may represent new therapeutic avenues to eradicate LSCs.


Subject(s)
Cell Adhesion Molecules/metabolism , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Stem Cell Niche/physiology , Animals , Bone Marrow/metabolism , Cell Adhesion/physiology , Hematopoietic Stem Cells/metabolism , Humans , Tumor Microenvironment/physiology
18.
Front Immunol ; 12: 766275, 2021.
Article in English | MEDLINE | ID: mdl-34858421

ABSTRACT

Knowledge about the hematopoietic niche has evolved considerably in recent years, in particular through in vitro analyzes, mouse models and the use of xenografts. Its complexity in the human bone marrow, in particular in a context of hematological malignancy, is more difficult to decipher by these strategies and could benefit from the knowledge acquired on the niches of solid tumors. Indeed, some common features can be suspected, since the bone marrow is a frequent site of solid tumor metastases. Recent research on solid tumors has provided very interesting information on the interactions between tumoral cells and their microenvironment, composed notably of mesenchymal, endothelial and immune cells. This review thus focuses on recent discoveries on tumor niches that could help in understanding hematopoietic niches, with special attention to 4 particular points: i) the heterogeneity of carcinoma/cancer-associated fibroblasts (CAFs) and mesenchymal stem/stromal cells (MSCs), ii) niche cytokines and chemokines, iii) the energy/oxidative metabolism and communication, especially mitochondrial transfer, and iv) the vascular niche through angiogenesis and endothelial plasticity. This review highlights actors and/or pathways of the microenvironment broadly involved in cancer processes. This opens avenues for innovative therapeutic opportunities targeting not only cancer stem cells but also their regulatory tumor niche(s), in order to improve current antitumor therapies.


Subject(s)
Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts , Cell Communication , Cytokines , Endothelium, Vascular , Humans , Mesenchymal Stem Cells , Neoplasms , Neovascularization, Pathologic
19.
J Exp Med ; 199(2): 221-9, 2004 Jan 19.
Article in English | MEDLINE | ID: mdl-14718516

ABSTRACT

Beta-catenin-mediated Wnt signaling has been suggested to be critically involved in hematopoietic stem cell maintenance and development of T and B cells in the immune system. Unexpectedly, here we report that inducible Cre-loxP-mediated inactivation of the beta-catenin gene in bone marrow progenitors does not impair their ability to self-renew and reconstitute all hematopoietic lineages (myeloid, erythroid, and lymphoid), even in competitive mixed chimeras. In addition, both thymocyte survival and antigen-induced proliferation of peripheral T cells is beta-catenin independent. In contrast to earlier reports, these data exclude an essential role for beta-catenin during hematopoiesis and lymphopoiesis.


Subject(s)
Cytoskeletal Proteins/physiology , Hematopoiesis/physiology , Lymphopoiesis/physiology , Trans-Activators/physiology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Cell Division , Chimera , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Female , Hematopoiesis/genetics , Integrases/genetics , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trans-Activators/deficiency , Trans-Activators/genetics , Wnt Proteins , beta Catenin
20.
Front Oncol ; 10: 606540, 2020.
Article in English | MEDLINE | ID: mdl-33489914

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) represents the malignant counterpart of bone marrow (BM) differentiating B cells and occurs most frequently in children. While new combinations of chemotherapeutic agents have dramatically improved the prognosis for young patients, disease outcome remains poor after relapse or in adult patients. This is likely due to heterogeneity of B-ALL response to treatment which relies not only on intrinsic properties of leukemic cells, but also on extrinsic protective cues transmitted by the tumor cell microenvironment. Alternatively, leukemic cells have the capacity to shape their microenvironment towards their needs. Most knowledge on the role of protective niches has emerged from the identification of mesenchymal and endothelial cells controlling hematopoietic stem cell self-renewal or B cell differentiation. In this review, we discuss the current knowledge about B-ALL protective niches and the development of therapies targeting the crosstalk between leukemic cells and their microenvironment.

SELECTION OF CITATIONS
SEARCH DETAIL