Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.980
Filtrer
1.
Compr Physiol ; 14(3): 5521-5579, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39109972

RÉSUMÉ

Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.


Sujet(s)
Tissu adipeux , Moelle osseuse , Humains , Animaux , Moelle osseuse/métabolisme , Moelle osseuse/physiologie , Tissu adipeux/métabolisme , Tissu adipeux/physiologie , Adipocytes/métabolisme , Adipocytes/physiologie , Adipogenèse/physiologie
2.
Exp Hematol ; 138: 104281, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39009278

RÉSUMÉ

Under stress hematopoiesis, previous studies have suggested the migration of hematopoietic stem cells (HSCs) from bone marrow (BM) to extramedullary sites such as the spleen. However, there is little direct evidence of HSC migration from the BM to the spleen. Here, we induced myeloablation via 5-fluorouracil (5-FU) and showed direct evidence of HSC migration from BM to spleen during hematopoietic regeneration via a photoconvertible fluorophore. Moreover, during steady state, HSCs preferentially migrated to BM rather than spleen, but during hematopoietic regeneration, HSCs preferred spleen as a migration site equivalently or greater. Furthermore, in the early phase, HSCs egressed from BM through the attenuated HSC retention. However, HSCs in the late phase gained significantly enhanced cell-autonomous motility, which was independent of chemotaxis. Collectively, HSC mobilization from BM, before the migration to the spleen, was dynamically changed from passive to active events during hematopoietic regeneration.


Sujet(s)
Mouvement cellulaire , Fluorouracil , Hématopoïèse , Cellules souches hématopoïétiques , Régénération , Rate , Animaux , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Souris , Rate/cytologie , Rate/métabolisme , Fluorouracil/pharmacologie , Mobilisation de cellules souches hématopoïétiques/méthodes , Souris de lignée C57BL , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/cytologie , Moelle osseuse/métabolisme , Moelle osseuse/physiologie
3.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999948

RÉSUMÉ

Extensive research has explored the functional correlation between stem cells and progenitor cells, particularly in blood. Hematopoietic stem cells (HSCs) can self-renew and regenerate tissues within the bone marrow, while stromal cells regulate tissue function. Recent studies have validated the role of mammalian stem cells within specific environments, providing initial empirical proof of this functional phenomenon. The interaction between bone and blood has always been vital to the function of the human body. It was initially proposed that during evolution, mammalian stem cells formed a complex relationship with the surrounding microenvironment, known as the niche. Researchers are currently debating the significance of molecular-level data to identify individual stromal cell types due to incomplete stromal cell mapping. Obtaining these data can help determine the specific activities of HSCs in bone marrow. This review summarizes key topics from previous studies on HSCs and their environment, discussing current and developing concepts related to HSCs and their niche in the bone marrow.


Sujet(s)
Moelle osseuse , Cellules souches hématopoïétiques , Niche de cellules souches , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Humains , Niche de cellules souches/physiologie , Animaux , Moelle osseuse/métabolisme , Moelle osseuse/physiologie , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/cytologie
4.
BMB Rep ; 57(8): 352-362, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38919014

RÉSUMÉ

Hematopoietic stem cell transplantation (HSCT) remains an indispensable therapeutic strategy for various hematological diseases. This review discusses the pivotal role of bone marrow (BM) niches in influencing the efficacy of HSCT and evaluates the current animal models, emphasizing their limitations and the need for alternative models. Traditional animal models, mainly murine xenograft, have provided significant insights, but due to species-specific differences, are often constrained from accurately mimicking human physiological responses. These limitations highlight the importance of developing alternative models that can more realistically replicate human hematopoiesis. Emerging models that include BM organoids and BM-on-a-chip microfluidic systems promise enhanced understanding of HSCT dynamics. These models aim to provide more accurate simulations of the human BM microenvironment, potentially leading to improved preclinical assessments and therapeutic outcomes. This review highlights the complexities of the BM niche, discusses the limitations of current models, and suggests directions for future research using advanced model systems. [BMB Reports 2024; 57(8): 352-362].


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Niche de cellules souches , Humains , Animaux , Transplantation de cellules souches hématopoïétiques/méthodes , Niche de cellules souches/physiologie , Moelle osseuse/physiologie , Moelle osseuse/métabolisme , Hématopoïèse/physiologie , Organoïdes , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/physiologie
5.
Gut Microbes ; 16(1): 2350784, 2024.
Article de Anglais | MEDLINE | ID: mdl-38727219

RÉSUMÉ

The gut microbiota constitutes a vast ecological system within the human body, forming a mutually interdependent entity with the host. In recent years, advancements in molecular biology technologies have provided a clearer understanding of the role of the gut microbiota. They not only influence the local immune status and metabolic functions of the host's intestinal tract but also impact the functional transformation of hematopoietic stem cells (HSCs) through the gut-blood axis. In this review, we will discuss the role of the gut microbiota in influencing hematopoiesis. We analyze the interactions between HSCs and other cellular components, with a particular emphasis on the direct functional regulation of HSCs by the gut microbiota and their indirect influence through cellular components in the bone marrow microenvironment. Additionally, we propose potential control targets for signaling pathways triggered by the gut microbiota to regulate hematopoietic function, filling crucial knowledge gaps in the development of this research field.


Sujet(s)
Microbiome gastro-intestinal , Hématopoïèse , Cellules souches hématopoïétiques , Hématopoïèse/physiologie , Microbiome gastro-intestinal/physiologie , Humains , Cellules souches hématopoïétiques/microbiologie , Animaux , Transduction du signal , Bactéries/métabolisme , Bactéries/classification , Bactéries/génétique , Tube digestif/microbiologie , Moelle osseuse/microbiologie , Moelle osseuse/physiologie
6.
Blood ; 144(1): 21-34, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38579285

RÉSUMÉ

ABSTRACT: Hematopoietic stem cells (HSCs) are instrumental for organismal survival because they are responsible for lifelong production of mature blood lineages in homeostasis and response to external stress. To fulfill their function, HSCs rely on reciprocal interactions with specialized tissue microenvironments, termed HSC niches. From embryonic development to advanced aging, HSCs transition through several hematopoietic organs in which they are supported by distinct extrinsic cues. Here, we describe recent discoveries on how HSC niches collectively adapt to ensure robust hematopoietic function during biological aging and after exposure to acute stress. We also discuss the latest strategies leveraging niche-derived signals to revert aging-associated phenotypes and enhance hematopoietic recovery after myeloablation.


Sujet(s)
Cellules souches hématopoïétiques , Niche de cellules souches , Stress physiologique , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/physiologie , Animaux , Humains , Niche de cellules souches/physiologie , Adaptation physiologique , Moelle osseuse/anatomopathologie , Moelle osseuse/métabolisme , Moelle osseuse/physiologie , Vieillissement/physiologie , Hématopoïèse/physiologie
7.
Eur J Radiol ; 172: 111359, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38325186

RÉSUMÉ

PURPOSE: Excess fat accumulation contributes significantly to metabolic dysfunction and diseases. This study aims to systematically compare the accuracy of commercially available Dixon techniques for quantification of fat fraction in liver, skeletal musculature, and vertebral bone marrow (BM) of healthy individuals, investigating biases and sex-specific influences. METHOD: 100 healthy White individuals (50 women) underwent abdominal MRI using two-point and multi-echo Dixon sequences. Fat fraction (FF), proton density fat fraction (PDFF) and T2* values were calculated for liver, paravertebral muscles (PVM) and vertebral BM (Th8-L5). Agreement and systematic deviations were assessed using linear correlation and Bland-Altman plots. RESULTS: High correlations between FF and PDFF were observed in liver (r = 0.98 for women; r = 0.96 for men), PVM (r = 0.92 for women; r = 0.93 for men) and BM (r = 0.97 for women; r = 0.95 for men). Relative deviations between FF and PDFF in liver (18.92 % for women; 13.32 % for men) and PVM (1.96 % for women; 11.62 % for men) were not significant. Relative deviations in BM were significant (38.13 % for women; 27.62 % for men). Bias correction using linear models reduced discrepancies. T2* times were significantly shorter in BM (8.72 ms for women; 7.26 ms for men) compared to PVM (13.45 ms for women; 13.62 ms for men) and liver (29.47 ms for women; 26.35 ms for men). CONCLUSION: While no significant differences were observed for liver and PVM, systematic errors in BM FF estimation using two-point Dixon imaging were observed. These discrepancies - mainly resulting from organ-specific T2* times - have to be considered when applying two-point Dixon approaches for assessment of fat content. As suitable correction tools, linear models could provide added value in large-scale epidemiological cohort studies. Sex-specific differences in T2* should be considered.


Sujet(s)
Moelle osseuse , Imagerie par résonance magnétique , Mâle , Humains , Femelle , Moelle osseuse/imagerie diagnostique , Moelle osseuse/physiologie , Imagerie par résonance magnétique/méthodes , Muscles squelettiques/imagerie diagnostique , Tissu adipeux/imagerie diagnostique , Foie/imagerie diagnostique
8.
Obes Rev ; 25(3): e13674, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38092420

RÉSUMÉ

Obesity has taken the face of a pandemic with less direct concern among the general population and scientific community. However, obesity is considered a low-grade systemic inflammation that impacts multiple organs. Chronic inflammation is also associated with different solid and blood cancers. In addition, emerging evidence demonstrates that individuals with obesity are at higher risk of developing blood cancers and have poorer clinical outcomes than individuals in a normal weight range. The bone marrow is critical for hematopoiesis, lymphopoiesis, and myelopoiesis. Therefore, it is vital to understand the mechanisms by which obesity-associated changes in BM adiposity impact leukemia development. BM adipocytes are critical to maintain homeostasis via different means, including immune regulation. However, obesity increases BM adiposity and creates a pro-inflammatory environment to upregulate clonal hematopoiesis and a leukemia-supportive environment. Obesity further alters lymphopoiesis and myelopoiesis via different mechanisms, which dysregulate myeloid and lymphoid immune cell functions mentioned in the text under different sequentially discussed sections. The altered immune cell function during obesity alters hematological malignancies and leukemia susceptibility. Therefore, obesity-induced altered BM adiposity, immune cell generation, and function impact an individual's predisposition and severity of leukemia, which should be considered a critical factor in leukemia patients.


Sujet(s)
Tumeurs hématologiques , Leucémies , Humains , Moelle osseuse/anatomopathologie , Moelle osseuse/physiologie , Adiposité , Obésité/complications , Obésité/anatomopathologie , Inflammation/anatomopathologie , Leucémies/étiologie , Leucémies/anatomopathologie , Tumeurs hématologiques/anatomopathologie
9.
Nat Cancer ; 4(8): 1193-1209, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37550517

RÉSUMÉ

Aging facilitates the expansion of hematopoietic stem cells (HSCs) carrying clonal hematopoiesis-related somatic mutations and the development of myeloid malignancies, such as myeloproliferative neoplasms (MPNs). While cooperating mutations can cause transformation, it is unclear whether distinct bone marrow (BM) HSC-niches can influence the growth and therapy response of HSCs carrying the same oncogenic driver. Here we found different BM niches for HSCs in MPN subtypes. JAK-STAT signaling differentially regulates CDC42-dependent HSC polarity, niche interaction and mutant cell expansion. Asymmetric HSC distribution causes differential BM niche remodeling: sinusoidal dilation in polycythemia vera and endosteal niche expansion in essential thrombocythemia. MPN development accelerates in a prematurely aged BM microenvironment, suggesting that the specialized niche can modulate mutant cell expansion. Finally, dissimilar HSC-niche interactions underpin variable clinical response to JAK inhibitor. Therefore, HSC-niche interactions influence the expansion rate and therapy response of cells carrying the same clonal hematopoiesis oncogenic driver.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Humains , Sujet âgé , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/thérapie , Syndromes myéloprolifératifs/anatomopathologie , Moelle osseuse/anatomopathologie , Moelle osseuse/physiologie , Cellules souches hématopoïétiques/anatomopathologie , Os et tissu osseux/anatomopathologie , Microenvironnement tumoral/génétique
10.
Nat Rev Endocrinol ; 19(11): 626-638, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37587198

RÉSUMÉ

Adipose tissue is a dynamic component of the bone marrow, regulating skeletal remodelling and secreting paracrine and endocrine factors that can affect haematopoiesis, as well as potentially nourishing the bone marrow during periods of stress. Bone marrow adipose tissue is regulated by multiple factors, but particularly nutrient status. In this Review, we examine how bone marrow adipocytes originate, their function in normal and pathological states and how bone marrow adipose tissue modulates whole-body homoeostasis through actions on bone cells, haematopoietic stem cells and extra-medullary adipocytes during nutritional challenges. We focus on both rodent models and human studies to help understand the unique marrow adipocyte, its response to the external nutrient environment and its effects on the skeleton. We finish by addressing some critical questions that to date remain unanswered.


Sujet(s)
Tissu adipeux , Cellules de la moelle osseuse , Moelle osseuse , Humains , Adipocytes/physiologie , Moelle osseuse/anatomopathologie , Moelle osseuse/physiologie , Cellules de la moelle osseuse/physiologie , Obésité/anatomopathologie , Perte de poids
11.
Cancer Commun (Lond) ; 43(6): 637-660, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37120719

RÉSUMÉ

BACKGROUND: Tumors possess incessant growth features, and expansion of their masses demands sufficient oxygen supply by red blood cells (RBCs). In adult mammals, the bone marrow (BM) is the main organ regulating hematopoiesis with dedicated manners. Other than BM, extramedullary hematopoiesis is discovered in various pathophysiological settings. However, whether tumors can contribute to hematopoiesis is completely unknown. Accumulating evidence shows that, in the tumor microenvironment (TME), perivascular localized cells retain progenitor cell properties and can differentiate into other cells. Here, we sought to better understand whether and how perivascular localized pericytes in tumors manipulate hematopoiesis. METHODS: To test if vascular cells can differentiate into RBCs, genome-wide expression profiling was performed using mouse-derived pericytes. Genetic tracing of perivascular localized cells employing NG2-CreERT2:R26R-tdTomato mouse strain was used to validate the findings in vivo. Fluorescence-activated cell sorting (FACS), single-cell sequencing, and colony formation assays were applied for biological studies. The production of erythroid differentiation-specific cytokine, erythropoietin (EPO), in TME was checked using quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA, magnetic-activated cell sorting and immunohistochemistry. To investigate BM function in tumor erythropoiesis, BM transplantation mouse models were employed. RESULTS: Genome-wide expression profiling showed that in response to platelet-derived growth factor subunit B (PDGF-B), neural/glial antigen 2 (NG2)+ perivascular localized cells exhibited hematopoietic stem and progenitor-like features and underwent differentiation towards the erythroid lineage. PDGF-B simultaneously targeted cancer-associated fibroblasts to produce high levels of EPO, a crucial hormone that necessitates erythropoiesis. FACS analysis using genetic tracing of NG2+ cells in tumors defined the perivascular localized cell-derived subpopulation of hematopoietic cells. Single-cell sequencing and colony formation assays validated the fact that, upon PDGF-B stimulation, NG2+ cells isolated from tumors acted as erythroblast progenitor cells, which were distinctive from the canonical BM hematopoietic stem cells. CONCLUSIONS: Our data provide a new concept of hematopoiesis within tumor tissues and novel mechanistic insights into perivascular localized cell-derived erythroid cells within TME. Targeting tumor hematopoiesis is a novel therapeutic concept for treating various cancers that may have profound impacts on cancer therapy.


Sujet(s)
Érythropoïèse , Tumeurs , Animaux , Souris , Moelle osseuse/physiologie , Différenciation cellulaire , Mammifères , Tumeurs/métabolisme , Péricytes , Microenvironnement tumoral
12.
Stem Cells ; 41(6): 541-551, 2023 06 15.
Article de Anglais | MEDLINE | ID: mdl-37061819

RÉSUMÉ

In mammals, post-injury repair and regenerative events rely predominantly on stem cell function. Stem cell transplantation has achieved considerable success in animals but remains unfavorable for humans because of the unavoidable drawbacks. Nevertheless, substantial evidence suggests the regenerative potential of endogenous stem cells can be improved for functional and structural recovery of tissue damage or in disease conditions. Endogenous stem cells are mostly quiescent under steady-state conditions and reside in their niche. Once faced with tissue injury, physiological and molecular changes within the niche or from distant tissues activate the migration, proliferation, and differentiation of stem cells, contributing to tissue repair. Tissue regeneration is augmented by artificially amplifying the factors that promote stem cell mobilization or enhance the homing of endogenous stem cells. This cell-free strategy, known as "in situ tissue regeneration," represents a safer and more efficient means to conduct tissue regeneration. Bone marrow (BM) is considered the central niche and main reservoir of many types of stem cells. These stem cells hold great therapeutic potential for the regeneration of multiple injured tissues. Herein, we review recent strategies for promoting in situ tissue regeneration through BM-derived stem cell mobilization or homing in animal models as well as in human trials. With the advancement in biomaterial engineering, chemoattractant signals combined with functionalized bioscaffolds have accomplished sustained activation of endogenous BM-derived stem cells that can be used as an attractive strategy for efficient in situ tissue regeneration.


Sujet(s)
Mobilisation de cellules souches hématopoïétiques , Transplantation de cellules souches hématopoïétiques , Animaux , Humains , Moelle osseuse/physiologie , Mouvement cellulaire/physiologie , Mammifères
13.
Elife ; 122023 03 06.
Article de Anglais | MEDLINE | ID: mdl-36876630

RÉSUMÉ

Hematopoiesis is regulated by the bone marrow (BM) stroma. However, cellular identities and functions of the different BM stromal elements in humans remain poorly defined. Based on single-cell RNA sequencing (scRNAseq), we systematically characterized the human non-hematopoietic BM stromal compartment and we investigated stromal cell regulation principles based on the RNA velocity analysis using scVelo and studied the interactions between the human BM stromal cells and hematopoietic cells based on ligand-receptor (LR) expression using CellPhoneDB. scRNAseq led to the identification of six transcriptionally and functionally distinct stromal cell populations. Stromal cell differentiation hierarchy was recapitulated based on RNA velocity analysis and in vitro proliferation capacities and differentiation potentials. Potential key factors that might govern the transition from stem and progenitor cells to fate-committed cells were identified. In situ localization analysis demonstrated that different stromal cells were localized in different niches in the bone marrow. In silico cell-cell communication analysis further predicted that different stromal cell types might regulate hematopoiesis through distinct mechanisms. These findings provide the basis for a comprehensive understanding of the cellular complexity of the human BM microenvironment and the intricate stroma-hematopoiesis crosstalk mechanisms, thus refining our current view on human hematopoietic niche organization.


Sujet(s)
Moelle osseuse , Cellules souches hématopoïétiques , Humains , Moelle osseuse/physiologie , Cellules souches hématopoïétiques/métabolisme , Niche de cellules souches/physiologie , Cellules de la moelle osseuse/métabolisme , Hématopoïèse/génétique , Analyse de séquence d'ARN , ARN/métabolisme
14.
Haematologica ; 108(8): 2205-2217, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-36861412

RÉSUMÉ

CD169, a specific marker for macrophages, is a member of the sialic acid-binding immunoglobulin-like lectin (Siglec) family which acts as an adhesion molecule implicated in cell-cell interaction via sialylated glycoconjugates. Although CD169+ macrophages have been found to participate in erythroblastic island (EBI) formation and support erythropoiesis under homeostasis and stress, the exact role of CD169 and its counter receptor in EBI remains unknown. Herein, we generated CD169-CreERT knock-in mice and investigated the function of CD169 in EBI formation and erythropoiesis using CD169-null mice. EBI formation was impaired in vitro by both blockade of CD169 using anti-CD169 antibody and deletion of CD169 on macrophages. Furthermore, CD43 expressed by early erythroblasts (EB) was identified as the counter receptor for CD169 in mediating the EBI formation via surface plasmon resonance and imaging flow cytometry. Interestingly, CD43 was proven to be a novel indicator of erythroid differentiation due to the progressive decrease of CD43 expression as EB mature. Although CD169-null mice did not display defects in bone marrow (BM) EBI formation in vivo, CD169 deficiency impeded BM erythroid differentiation probably via CD43 under stress erythropoiesis, in concert with the role of CD169 recombinant protein in hemin-induced K562 erythroid differentiation. These findings have shed light on the role of CD169 in EBI under steady and stress erythropoiesis through binding with its counter receptor CD43, suggesting that CD169-CD43 interaction might be a promising therapeutic target for erythroid disorders.


Sujet(s)
Moelle osseuse , Érythroblastes , Animaux , Souris , Érythroblastes/métabolisme , Moelle osseuse/physiologie , Érythropoïèse , Macrophages/métabolisme , Différenciation cellulaire , Souris knockout
15.
Curr Opin Hematol ; 30(3): 80-85, 2023 05 01.
Article de Anglais | MEDLINE | ID: mdl-36718814

RÉSUMÉ

PURPOSE OF REVIEW: Terminal erythroid differentiation occurs in specialized niches called erythroblastic islands. Since their discovery in 1958, these niches have been described as a central macrophage surrounded by differentiating erythroblasts. Here, we review the recent advances made in the characterization of these islands and the role they could play in anaemia of inflammation. RECENT FINDINGS: The utilization of multispectral imaging flow cytometry (flow cytometry with microscopy) has enabled for a more precise characterization of the niche that revealed the presence of maturing granulocytes in close contact with the central macrophage. These erythromyeloblastic islands (EMBIs) can adapt depending on the peripheral needs. Indeed, during inflammation wherein inflammatory cytokines limit erythropoiesis and promote granulopoiesis, EMBIs present altered structures with increased maturing granulocytes and decreased erythroid precursors. SUMMARY: Regulation of the structure and function of the EMBI in the bone marrow emerges as a potential player in the pathophysiology of acute and chronic inflammation and its associated anaemia.


Sujet(s)
Anémie , Moelle osseuse , Humains , Moelle osseuse/physiologie , Érythroblastes , Érythropoïèse/physiologie , Anémie/étiologie , Inflammation
16.
J Exp Med ; 220(4)2023 04 03.
Article de Anglais | MEDLINE | ID: mdl-36719648

RÉSUMÉ

Emergency myelopoiesis (EM) is a hematopoietic response against systemic infections that quickly supplies innate immune cells. As lymphopoiesis is strongly suppressed during EM, the role of lymphocytes in that process has not received much attention. Here, we found that myeloid-like B cells (M-B cells), which express myeloid markers, emerge in the bone marrow (BM) after the induction of EM. M-B cells were mainly derived from pre-B cells and preferentially expressed IL-10, which directly stimulates hematopoietic progenitors to enhance their survival and myeloid-biased differentiation. Indeed, lacking IL-10 in B cells, blocking IL-10 in the BM with a neutralizing antibody, and deleting the IL-10 receptor in hematopoietic progenitors significantly suppressed EM, which failed to clear microbes in a cecal ligation and puncture model. Thus, a distinct B cell subset generated during infection plays a pivotal role in boosting EM, which suggests the on-demand reinforcement of EM by adaptive immune cells.


Sujet(s)
Lymphocytes B , Interleukine-10 , Myélopoïèse , Moelle osseuse/physiologie , Cellules de la moelle osseuse , Hématopoïèse , Cellules myéloïdes
17.
J Foot Ankle Surg ; 62(1): 75-79, 2023.
Article de Anglais | MEDLINE | ID: mdl-35659478

RÉSUMÉ

Bone marrow stimulation (BMS) is indicated for patients who have symptomatic osteochondral lesions of the talus (OLT). Despite differences in ankle biomechanics and cartilage morphology between men and women, there is scant evidence examining whether these differences affect surgical outcomes. The purpose of this study was to compare the outcomes in men and women following BMS for OLTs. A retrospective analysis comparing female and male patients treated with BMS for OLT between 2007 and 2015 was performed. Clinical outcomes were evaluated using the Foot and Ankle Outcome Scores (FAOS) and Short-Form 12 (SF-12). Magnetic resonance imaging at final follow-up was evaluated with the modified magnetic resonance observation of cartilage repair tissue score. Thirty-one females and 38 males were included. In female patients, the mean FAOS pain score improved from 60 ± 16 preoperatively to 84 ± 8.9 at 1- to 2-year follow-up (p < .01), and then decreased to 80±13 at final follow-up at 3-4 years. In male patients, the mean FAOS pain score improved from 65±17 preoperatively to 83±9.2 at 1-2 year follow-up (p < .01), and then decreased to 76±14.6 at final follow-up at 3-4 years. Lateral lesions were more common in male patients. Medial lesions were more common in female patients. The outcomes following BMS in both female and male patients were good with no significant differences at short-term follow-up. FAOS scores in male patients were more likely to decrease after 1 to 2 years postsurgery, implying a possibly faster decline than in female patients.


Sujet(s)
Cartilage articulaire , Fractures articulaires , Talus , Humains , Mâle , Femelle , Études rétrospectives , Moelle osseuse/imagerie diagnostique , Moelle osseuse/physiologie , Talus/imagerie diagnostique , Talus/chirurgie , Talus/traumatismes , Facteurs sexuels , Imagerie par résonance magnétique , Douleur , Résultat thérapeutique , Cartilage articulaire/imagerie diagnostique , Cartilage articulaire/chirurgie , Cartilage articulaire/anatomopathologie
18.
Nat Commun ; 13(1): 7657, 2022 12 10.
Article de Anglais | MEDLINE | ID: mdl-36496394

RÉSUMÉ

Innate and adaptive immune cells participate in the homeostatic regulation of hematopoietic stem cells (HSCs). Here, we interrogate the contribution of myeloid cells, the most abundant cell type in the mammalian bone marrow, in a clinically relevant mouse model of neutropenia. Long-term genetic depletion of neutrophils and eosinophils results in activation of multipotent progenitors but preservation of HSCs. Depletion of myeloid cells abrogates HSC expansion, loss of serial repopulation and lymphoid reconstitution capacity and remodeling of HSC niches, features previously associated with hematopoietic aging. This is associated with mitigation of interferon signaling in both HSCs and their niches via reduction of NK cell number and activation. These data implicate myeloid cells in the functional decline of hematopoiesis, associated with activation of interferon signaling via a putative neutrophil-NK cell axis. Innate immunity may thus come at the cost of system deterioration through enhanced chronic inflammatory signaling to stem cells and their niches.


Sujet(s)
Hématopoïèse , Cellules souches hématopoïétiques , Souris , Animaux , Cellules souches hématopoïétiques/métabolisme , Cellules myéloïdes , Moelle osseuse/physiologie , Interférons/métabolisme , Différenciation cellulaire , Mammifères
19.
Elife ; 112022 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-36178184

RÉSUMÉ

Generation of bone marrow (BM) from embryonic stem cells (ESCs) promises to accelerate the development of future cell therapies for life-threatening disorders. However, such approach is limited by technical challenges to produce a mixture of functional BM progenitor cells able to replace all hematopoietic cell lineages. Herein, we used blastocyst complementation to simultaneously produce BM cell lineages from mouse ESCs in a rat. Based on fluorescence-activated cell sorting analysis and single-cell RNA sequencing, mouse ESCs differentiated into multiple hematopoietic and stromal cell types that were indistinguishable from normal mouse BM cells based on gene expression signatures and cell surface markers. Receptor-ligand interactions identified Cxcl12-Cxcr4, Lama2-Itga6, App-Itga6, Comp-Cd47, Col1a1-Cd44, and App-Il18rap as major signaling pathways between hematopoietic progenitors and stromal cells. Multiple hematopoietic progenitors, including hematopoietic stem cells (HSCs) in mouse-rat chimeras derived more efficiently from mouse ESCs, whereas chondrocytes predominantly derived from rat cells. In the dorsal aorta and fetal liver of mouse-rat chimeras, mouse HSCs emerged and expanded faster compared to endogenous rat cells. Sequential BM transplantation of ESC-derived cells from mouse-rat chimeras rescued lethally irradiated syngeneic mice and demonstrated long-term reconstitution potential of donor HSCs. Altogether, a fully functional BM was generated from mouse ESCs using rat embryos as 'bioreactors'.


Sujet(s)
Moelle osseuse , Transplantation de cellules souches hématopoïétiques , Souris , Animaux , Rats , Moelle osseuse/physiologie , Antigènes CD47 , Chimère , Ligands , Cellules souches embryonnaires , Cellules de la moelle osseuse
20.
Nat Neurosci ; 25(5): 555-560, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35301477

RÉSUMÉ

It remains unclear how immune cells from skull bone marrow niches are recruited to the meninges. Here we report that cerebrospinal fluid (CSF) accesses skull bone marrow via dura-skull channels, and CSF proteins signal onto diverse cell types within the niches. After spinal cord injury, CSF-borne cues promote myelopoiesis and egress of myeloid cells into meninges. This reveals a mechanism of CNS-to-bone-marrow communication via CSF that regulates CNS immune responses.


Sujet(s)
Moelle osseuse , Crâne , Moelle osseuse/physiologie , Liquide cérébrospinal , Tête , Méninges , Cellules myéloïdes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE