Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.851
Filtrer
1.
Nat Commun ; 15(1): 4965, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862518

RÉSUMÉ

Sickle cell disease is a devastating blood disorder that originates from a single point mutation in the HBB gene coding for hemoglobin. Here, we develop a GMP-compatible TALEN-mediated gene editing process enabling efficient HBB correction via a DNA repair template while minimizing risks associated with HBB inactivation. Comparing viral versus non-viral DNA repair template delivery in hematopoietic stem and progenitor cells in vitro, both strategies achieve comparable HBB correction and result in over 50% expression of normal adult hemoglobin in red blood cells without inducing ß-thalassemic phenotype. In an immunodeficient female mouse model, transplanted cells edited with the non-viral strategy exhibit higher engraftment and gene correction levels compared to those edited with the viral strategy. Transcriptomic analysis reveals that non-viral DNA repair template delivery mitigates P53-mediated toxicity and preserves high levels of long-term hematopoietic stem cells. This work paves the way for TALEN-based autologous gene therapy for sickle cell disease.


Sujet(s)
Drépanocytose , Édition de gène , Thérapie génétique , Cellules souches hématopoïétiques , Nucléases effectrices de type activateur de transcription , Drépanocytose/thérapie , Drépanocytose/génétique , Édition de gène/méthodes , Animaux , Cellules souches hématopoïétiques/métabolisme , Humains , Femelle , Souris , Thérapie génétique/méthodes , Nucléases effectrices de type activateur de transcription/métabolisme , Nucléases effectrices de type activateur de transcription/génétique , Transplantation de cellules souches hématopoïétiques , Globines bêta/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Réparation de l'ADN , Mutation , bêta-Thalassémie/thérapie , bêta-Thalassémie/génétique , Modèles animaux de maladie humaine , Techniques de transfert de gènes
2.
Stem Cell Res Ther ; 15(1): 164, 2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38853275

RÉSUMÉ

BACKGROUND: Transplantation of CD34+ hematopoietic stem and progenitor cells (HSPC) into immunodeficient mice is an established method to generate humanized mice harbouring a human immune system. Different sources and methods for CD34+ isolation have been employed by various research groups, resulting in customized models that are difficult to compare. A more detailed characterization of CD34+ isolates is needed for a better understanding of engraftable hematopoietic and potentially non-hematopoietic cells. Here we have performed a direct comparison of CD34+ isolated from cord blood (CB-CD34+) or fetal liver (FL-CD34+ and FL-CD34+CD14-) and their engraftment into immunocompromised NOD/Shi-scid Il2rgnull (NOG) mice. METHODS: NOG mice were transplanted with either CB-CD34+, FL-CD34+ or FL-CD34+CD14- to generate CB-NOG, FL-NOG and FL-CD14--NOG, respectively. After 15-20 weeks, the mice were sacrificed and human immune cell reconstitution was assessed in blood and several organs. Liver sections were pathologically assessed upon Haematoxylin and Eosin staining. To assess the capability of allogenic tumor rejection in CB- vs. FL-reconstituted mice, animals were subcutaneously engrafted with an HLA-mismatched melanoma cell line. Tumor growth was assessed by calliper measurements and a Luminex-based assay was used to compare the cytokine/chemokine profiles. RESULTS: We show that CB-CD34+ are a uniform population of HSPC that reconstitute NOG mice more rapidly than FL-CD34+ due to faster B cell development. However, upon long-term engraftment, FL-NOG display increased numbers of neutrophils, dendritic cells and macrophages in multiple tissues. In addition to HSPC, FL-CD34+ isolates contain non-hematopoietic CD14+ endothelial cells that enhance the engraftment of the human immune system in FL-NOG mice. We demonstrate that these CD14+CD34+ cells are capable of reconstituting Factor VIII-producing liver sinusoidal endothelial cells (LSEC) in FL-NOG. However, CD14+CD34+ also contribute to hepatic sinusoidal dilatation and immune cell infiltration, which may culminate in a graft-versus-host disease (GVHD) pathology upon long-term engraftment. Finally, using an HLA-A mismatched CDX melanoma model, we show that FL-NOG, but not CB-NOG, can mount a graft-versus-tumor (GVT) response resulting in tumor rejection. CONCLUSION: Our results highlight important phenotypical and functional differences between CB- and FL-NOG and reveal FL-NOG as a potential model to study hepatic sinusoidal dilatation and mechanisms of GVT.


Sujet(s)
Antigènes CD34 , Foie , Animaux , Humains , Antigènes CD34/métabolisme , Souris , Foie/métabolisme , Foie/anatomopathologie , Souris de lignée NOD , Transplantation de cellules souches hématopoïétiques , Souris SCID , Progéniteurs endothéliaux/métabolisme , Progéniteurs endothéliaux/cytologie , Progéniteurs endothéliaux/transplantation , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Sang foetal/cytologie , Mélanome/anatomopathologie , Mélanome/immunologie
3.
J Cell Biol ; 223(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-38874393

RÉSUMÉ

Hematopoietic stem cells (HSCs) continuously replenish mature blood cells with limited lifespans. To maintain the HSC compartment while ensuring output of differentiated cells, HSCs undergo asymmetric cell division (ACD), generating two daughter cells with different fates: one will proliferate and give rise to the differentiated cells' progeny, and one will return to quiescence to maintain the HSC compartment. A balance between MEK/ERK and mTORC1 pathways is needed to ensure HSC homeostasis. Here, we show that activation of these pathways is spatially segregated in premitotic HSCs and unequally inherited during ACD. A combination of genetic and chemical perturbations shows that an ERK-dependent mechanism determines the balance between pathways affecting polarity, proliferation, and metabolism, and thus determines the frequency of asymmetrically dividing HSCs. Our data identify druggable targets that modulate HSC fate determination at the level of asymmetric division.


Sujet(s)
Division cellulaire asymétrique , Cellules souches hématopoïétiques , Animaux , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Différenciation cellulaire , Transduction du signal , Prolifération cellulaire , Lignage cellulaire , Souris , Système de signalisation des MAP kinases , Souris de lignée C57BL , Polarité de la cellule
4.
Sci Rep ; 14(1): 14080, 2024 06 18.
Article de Anglais | MEDLINE | ID: mdl-38890442

RÉSUMÉ

Familial platelet disorder with associated myeloid malignancies (FPDMM) is an autosomal dominant disease caused by heterozygous germline mutations in RUNX1. It is characterized by thrombocytopenia, platelet dysfunction, and a predisposition to hematological malignancies. Although FPDMM is a precursor for diseases involving abnormal DNA methylation, the DNA methylation status in FPDMM remains unknown, largely due to a lack of animal models and challenges in obtaining patient-derived samples. Here, using genome editing techniques, we established two lines of human induced pluripotent stem cells (iPSCs) with different FPDMM-mimicking heterozygous RUNX1 mutations. These iPSCs showed defective differentiation of hematopoietic progenitor cells (HPCs) and megakaryocytes (Mks), consistent with FPDMM. The FPDMM-mimicking HPCs showed DNA methylation patterns distinct from those of wild-type HPCs, with hypermethylated regions showing the enrichment of ETS transcription factor (TF) motifs. We found that the expression of FLI1, an ETS family member, was significantly downregulated in FPDMM-mimicking HPCs with a RUNX1 transactivation domain (TAD) mutation. We demonstrated that FLI1 promoted binding-site-directed DNA demethylation, and that overexpression of FLI1 restored their megakaryocytic differentiation efficiency and hypermethylation status. These findings suggest that FLI1 plays a crucial role in regulating DNA methylation and correcting defective megakaryocytic differentiation in FPDMM-mimicking HPCs with a RUNX1 TAD mutation.


Sujet(s)
Différenciation cellulaire , Sous-unité alpha 2 du facteur CBF , Méthylation de l'ADN , Cellules souches pluripotentes induites , Mégacaryocytes , Mutation , Protéine proto-oncogène c-fli-1 , Sous-unité alpha 2 du facteur CBF/génétique , Sous-unité alpha 2 du facteur CBF/métabolisme , Humains , Mégacaryocytes/métabolisme , Protéine proto-oncogène c-fli-1/génétique , Protéine proto-oncogène c-fli-1/métabolisme , Différenciation cellulaire/génétique , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Anomalies des plaquettes/génétique , Anomalies des plaquettes/métabolisme , Anomalies des plaquettes/anatomopathologie , Activation de la transcription , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Leucémie aigüe myéloïde , Troubles héréditaires de la coagulation sanguine
5.
Nature ; 630(8016): 412-420, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839950

RÉSUMÉ

The processes that govern human haematopoietic stem cell (HSC) self-renewal and engraftment are poorly understood and challenging to recapitulate in culture to reliably expand functional HSCs1-3. Here we identify MYC target 1 (MYCT1; also known as MTLC) as a crucial human HSC regulator that moderates endocytosis and environmental sensing in HSCs. MYCT1 is selectively expressed in undifferentiated human haematopoietic stem and progenitor cells (HSPCs) and endothelial cells but becomes markedly downregulated during HSC culture. Lentivirus-mediated knockdown of MYCT1 prevented human fetal liver and cord blood (CB) HSPC expansion and engraftment. By contrast, restoring MYCT1 expression improved the expansion and engraftment of cultured CB HSPCs. Single-cell RNA sequencing of human CB HSPCs in which MYCT1 was knocked down or overexpressed revealed that MYCT1 governs important regulatory programmes and cellular properties essential for HSC stemness, such as ETS factor expression and low mitochondrial activity. MYCT1 is localized in the endosomal membrane in HSPCs and interacts with vesicle trafficking regulators and signalling machinery. MYCT1 loss in HSPCs led to excessive endocytosis and hyperactive signalling responses, whereas restoring MYCT1 expression balanced culture-induced endocytosis and dysregulated signalling. Moreover, sorting cultured CB HSPCs on the basis of lowest endocytosis rate identified HSPCs with preserved MYCT1 expression and MYCT1-regulated HSC stemness programmes. Our work identifies MYCT1-moderated endocytosis and environmental sensing as essential regulatory mechanisms required to preserve human HSC stemness. Our data also pinpoint silencing of MYCT1 as a cell-culture-induced vulnerability that compromises human HSC expansion.


Sujet(s)
Auto-renouvellement cellulaire , Cellules souches hématopoïétiques , Protéines nucléaires , Animaux , Femelle , Humains , Mâle , Souris , Cellules cultivées , Endocytose , Endosomes/métabolisme , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Sang foetal/cytologie , Techniques de knock-down de gènes , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Foie/cytologie , Foie/métabolisme , Foie/embryologie , Mitochondries/métabolisme , Protéines nucléaires/métabolisme , Transduction du signal , Protéines proto-oncogènes c-ets/génétique , Protéines proto-oncogènes c-ets/métabolisme , Analyse de l'expression du gène de la cellule unique
6.
Nat Commun ; 15(1): 5272, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902243

RÉSUMÉ

While myelodysplastic syndromes with del(5q) (del(5q) MDS) comprises a well-defined hematological subgroup, the molecular basis underlying its origin remains unknown. Using single cell RNA-seq (scRNA-seq) on CD34+ progenitors from del(5q) MDS patients, we have identified cells harboring the deletion, characterizing the transcriptional impact of this genetic insult on disease pathogenesis and treatment response. Interestingly, both del(5q) and non-del(5q) cells present similar transcriptional lesions, indicating that all cells, and not only those harboring the deletion, may contribute to aberrant hematopoietic differentiation. However, gene regulatory network (GRN) analyses reveal a group of regulons showing aberrant activity that could trigger altered hematopoiesis exclusively in del(5q) cells, pointing to a more prominent role of these cells in disease phenotype. In del(5q) MDS patients achieving hematological response upon lenalidomide treatment, the drug reverts several transcriptional alterations in both del(5q) and non-del(5q) cells, but other lesions remain, which may be responsible for potential future relapses. Moreover, lack of hematological response is associated with the inability of lenalidomide to reverse transcriptional alterations. Collectively, this study reveals transcriptional alterations that could contribute to the pathogenesis and treatment response of del(5q) MDS.


Sujet(s)
Antigènes CD34 , Délétion de segment de chromosome , Chromosomes humains de la paire 5 , Cellules souches hématopoïétiques , Lénalidomide , Syndromes myélodysplasiques , Analyse sur cellule unique , Humains , Lénalidomide/pharmacologie , Lénalidomide/usage thérapeutique , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/anatomopathologie , Syndromes myélodysplasiques/métabolisme , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Antigènes CD34/métabolisme , Chromosomes humains de la paire 5/génétique , Mâle , Femelle , Sujet âgé , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Hématopoïèse/effets des médicaments et des substances chimiques , Hématopoïèse/génétique , Transcriptome , Sujet âgé de 80 ans ou plus , RNA-Seq , Analyse de profil d'expression de gènes
7.
Sci Rep ; 14(1): 14187, 2024 06 20.
Article de Anglais | MEDLINE | ID: mdl-38902328

RÉSUMÉ

Mononuclear phagocytes (MNP), including macrophages and dendritic cells form an essential component of primary responses to environmental hazards and toxic exposures. This is particularly important in disease conditions such as asthma and allergic airway disease, where many different cell types are present. In this study, we differentiated CD34+ haematopoietic stem cells towards different populations of MNP in an effort to understand how different cell subtypes present in inflammatory disease microenvironments respond to the common allergen house dust mite (HDM). Using single cell mRNA sequencing, we demonstrate that macrophage subtypes MCSPP1+ and MLCMARCO+ display different patterns of gene expression after HDM challenge, noted especially for the chemokines CXCL5, CXCL8, CCL5 and CCL15. MLCCD206Hi alternatively activated macrophages displayed the greatest changes in expression, while neutrophil and monocyte populations did not respond. Further work investigated how pollutant diesel exhaust particles could modify these transcriptional responses and revealed that CXC but not CC type chemokines were further upregulated. Through the use of diesel particles with adsorbed material removed, we suggest that soluble pollutants on these particles are the active constituents responsible for the modifying effects on HDM. This study highlights that environmental exposures may influence tissue responses dependent on which MNP cell type is present, and that these should be considerations when modelling such events in vitro. Understanding the nuanced responsiveness of different immune cell types to allergen and pollutant exposure also contributes to a better understanding of how these exposures influence the development and exacerbation of human disease.


Sujet(s)
Pyroglyphidae , Animaux , Pyroglyphidae/immunologie , Humains , Phagocytes/métabolisme , Phagocytes/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Allergènes/immunologie , Emissions des véhicules/toxicité , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
8.
J Transl Med ; 22(1): 526, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38822352

RÉSUMÉ

BACKGROUND: Neutrophils are granulocytes with essential antimicrobial effector functions and short lifespans. During infection or sterile inflammation, emergency granulopoiesis leads to release of immature neutrophils from the bone marrow, serving to boost circulating neutrophil counts. Steady state and emergency granulopoiesis are incompletely understood, partly due to a lack of genetically amenable models of neutrophil development. METHODS: We optimised a method for ex vivo production of human neutrophils from CD34+ haematopoietic progenitors. Using flow cytometry, we phenotypically compared cultured neutrophils with native neutrophils from donors experiencing emergency granulopoiesis, and steady state neutrophils from non-challenged donors. We carry out functional and proteomic characterisation of cultured neutrophils and establish genome editing of progenitors. RESULTS: We obtain high yields of ex vivo cultured neutrophils, which phenotypically resemble immature neutrophils released into the circulation during emergency granulopoiesis. Cultured neutrophils have similar rates of ROS production and bacterial killing but altered degranulation, cytokine release and antifungal activity compared to mature neutrophils isolated from peripheral blood. These differences are likely due to incomplete synthesis of granule proteins, as demonstrated by proteomic analysis. CONCLUSION: Ex vivo cultured neutrophils are genetically tractable via genome editing of precursors and provide a powerful model system for investigating the properties and behaviour of immature neutrophils.


Sujet(s)
Antigènes CD34 , Granulocytes neutrophiles , Humains , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/cytologie , Antigènes CD34/métabolisme , Cellules cultivées , Espèces réactives de l'oxygène/métabolisme , Protéomique , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Édition de gène , Dégranulation cellulaire , Cellules souches/métabolisme , Cellules souches/cytologie , Cytokines/métabolisme , Phénotype
9.
Life Sci Alliance ; 7(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38830768

RÉSUMÉ

Hematopoietic stem cells and multipotential progenitors emerge in multiple, overlapping waves of fetal development. Some of these populations seed the bone marrow and sustain adult B- and T-cell development long-term after birth. However, others are present transiently, but whether they are vestigial or generate B and T cells that contribute to the adult immune system is not well understood. We now report that transient fetal progenitors distinguished by expression of low levels of the PU.1 transcription factor generated activated and memory T and B cells that colonized and were maintained in secondary lymphoid tissues. These included the small and large intestines, where they may contribute to the maintenance of gut homeostasis through at least middle age. At least some of the activated/memory cells may have been the progeny of B-1 and marginal zone B cells, as transient PU.1low fetal progenitors efficiently generated those populations. Taken together, our data demonstrate the potential of B- and T-cell progeny of transient PU.1low fetal progenitors to make an early and long-term contribution to the adult immune system.


Sujet(s)
Lymphocytes B , Protéines proto-oncogènes , Lymphocytes T , Transactivateurs , Transactivateurs/métabolisme , Transactivateurs/génétique , Animaux , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Souris , Lymphocytes B/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/cytologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/cytologie , Souris de lignée C57BL , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Différenciation cellulaire/immunologie , Femelle , Foetus/cytologie , Cellules souches foetales/métabolisme , Cellules souches foetales/cytologie
10.
Cells ; 13(12)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38920667

RÉSUMÉ

Hematopoietic stem cell (HSC) transduction has undergone remarkable advancements in recent years, revolutionizing the landscape of gene therapy specifically for inherited hematologic disorders. The evolution of viral vector-based transduction technologies, including retroviral and lentiviral vectors, has significantly enhanced the efficiency and specificity of gene delivery to HSCs. Additionally, the emergence of small molecules acting as transduction enhancers has addressed critical barriers in HSC transduction, unlocking new possibilities for therapeutic intervention. Furthermore, the advent of gene editing technologies, notably CRISPR-Cas9, has empowered precise genome modification in HSCs, paving the way for targeted gene correction. These striking progresses have led to the clinical approval of medicinal products based on engineered HSCs with impressive therapeutic benefits for patients. This review provides a comprehensive overview of the collective progress in HSC transduction via viral vectors for gene therapy with a specific focus on transduction enhancers, highlighting the latest key developments, challenges, and future directions towards personalized and curative treatments.


Sujet(s)
Thérapie génétique , Vecteurs génétiques , Cellules souches hématopoïétiques , Transduction génétique , Humains , Thérapie génétique/méthodes , Cellules souches hématopoïétiques/métabolisme , Animaux , Transplantation de cellules souches hématopoïétiques , Édition de gène/méthodes
12.
Development ; 151(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38940293

RÉSUMÉ

Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.


Sujet(s)
Différenciation cellulaire , Hématopoïèse , Cellules souches hématopoïétiques , Danio zébré , Animaux , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Souris , Différenciation cellulaire/effets des médicaments et des substances chimiques , Hématopoïèse/effets des médicaments et des substances chimiques , Récepteurs Notch/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Corps embryoïdes/cytologie , Corps embryoïdes/effets des médicaments et des substances chimiques , Corps embryoïdes/métabolisme , Cils vibratiles/métabolisme , Cils vibratiles/effets des médicaments et des substances chimiques , Blastomères/cytologie , Blastomères/métabolisme , Blastomères/effets des médicaments et des substances chimiques , Cellules cultivées
13.
Biol Open ; 13(6)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38828842

RÉSUMÉ

Most hematological malignancies are associated with reduced expression of one or more components of the Endosomal Sorting Complex Required for Transport (ESCRT). However, the roles of ESCRT in stem cell and progenitor maintenance are not resolved. Parsing signaling pathways in relation to the canonical role of ESCRT poses a challenge. The Drosophila hematopoietic organ, the larval lymph gland, provides a path to dissect the roles of cellular trafficking pathways such as ESCRT in blood development and maintenance. Drosophila has 13 core ESCRT components. Knockdown of individual ESCRTs showed that only Vps28 and Vp36 were required in all lymph gland progenitors. Using the well-conserved ESCRT-II complex as an example of the range of phenotypes seen upon ESCRT depletion, we show that ESCRTs have cell-autonomous as well as non-autonomous roles in progenitor maintenance and differentiation. ESCRT depletion also sensitized posterior lobe progenitors to respond to immunogenic wasp infestation. We also identify key heterotypic roles for ESCRT in position-dependent control of Notch activation to suppress crystal cell differentiation. Our study shows that the cargo sorting machinery determines the identity of progenitors and their adaptability to the dynamic microenvironment. These mechanisms for control of cell fate may tailor developmental diversity in multiple contexts.


Sujet(s)
Complexes de tri endosomique requis pour le transport , Animaux , Complexes de tri endosomique requis pour le transport/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Lignage cellulaire , Différenciation cellulaire/génétique , Drosophila , Transduction du signal , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Immunité
14.
BMC Biol ; 22(1): 143, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937802

RÉSUMÉ

BACKGROUND: The endothelial-to-hematopoietic transition (EHT) process during definitive hematopoiesis is highly conserved in vertebrates. Stage-specific expression of transposable elements (TEs) has been detected during zebrafish EHT and may promote hematopoietic stem cell (HSC) formation by activating inflammatory signaling. However, little is known about how TEs contribute to the EHT process in human and mouse. RESULTS: We reconstructed the single-cell EHT trajectories of human and mouse and resolved the dynamic expression patterns of TEs during EHT. Most TEs presented a transient co-upregulation pattern along the conserved EHT trajectories, coinciding with the temporal relaxation of epigenetic silencing systems. TE products can be sensed by multiple pattern recognition receptors, triggering inflammatory signaling to facilitate HSC emergence. Interestingly, we observed that hypoxia-related signals were enriched in cells with higher TE expression. Furthermore, we constructed the hematopoietic cis-regulatory network of accessible TEs and identified potential TE-derived enhancers that may boost the expression of specific EHT marker genes. CONCLUSIONS: Our study provides a systematic vision of how TEs are dynamically controlled to promote the hematopoietic fate decisions through transcriptional and cis-regulatory networks, and pre-train the immunity of nascent HSCs.


Sujet(s)
Éléments transposables d'ADN , Hématopoïèse , Cellules souches hématopoïétiques , Analyse sur cellule unique , Animaux , Éléments transposables d'ADN/génétique , Analyse sur cellule unique/méthodes , Souris , Hématopoïèse/génétique , Humains , Cellules souches hématopoïétiques/métabolisme , Cellules endothéliales/métabolisme
15.
J Control Release ; 371: 386-405, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38844177

RÉSUMÉ

Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.


Sujet(s)
Protéine morphogénétique osseuse de type 4 , Différenciation cellulaire , Gélatine , Cellules souches hématopoïétiques , Cellules souches pluripotentes induites , Sphéroïdes de cellules , Facteur de croissance des cellules souches , Protéine morphogénétique osseuse de type 4/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Facteur de croissance des cellules souches/métabolisme , Gélatine/composition chimique , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Animaux , Humains , Souris
16.
Nat Commun ; 15(1): 5090, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918373

RÉSUMÉ

The development of haematopoiesis involves the coordinated action of numerous genes, some of which are implicated in haematological malignancies. However, the biological function of many genes remains elusive and unknown functional genes are likely to remain to be uncovered. Here, we report a previously uncharacterised gene in haematopoiesis, identified by screening mutant embryonic stem cells. The gene, 'attenuated haematopoietic development (Ahed)', encodes a nuclear protein. Conditional knockout (cKO) of Ahed results in anaemia from embryonic day 14.5 onward, leading to prenatal demise. Transplantation experiments demonstrate the incapacity of Ahed-deficient haematopoietic cells to reconstitute haematopoiesis in vivo. Employing a tamoxifen-inducible cKO model, we further reveal that Ahed deletion impairs the intrinsic capacity of haematopoietic cells in adult mice. Ahed deletion affects various pathways, and published databases present cancer patients with somatic mutations in Ahed. Collectively, our findings underscore the fundamental roles of Ahed in lifelong haematopoiesis, implicating its association with malignancies.


Sujet(s)
Hématopoïèse , Souris knockout , Animaux , Hématopoïèse/génétique , Souris , Humains , Femelle , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Souris de lignée C57BL , Mutation , Anémie/génétique , Mâle , Cellules souches embryonnaires/métabolisme
18.
Genes (Basel) ; 15(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38927642

RÉSUMÉ

Chronic granulomatous disease (CGD) is an inherited immunodeficiency disease mainly caused by mutations in the X-linked CYBB gene that abrogate reactive oxygen species (ROS) production in phagocytes and microbial defense. Gene repair using the CRISPR/Cas9 system in hematopoietic stem and progenitor cells (HSPCs) is a promising technology for therapy for CGD. To support the establishment of efficient and safe gene therapies for CGD, we generated a mouse model harboring a patient-derived mutation in the CYBB gene. Our CybbC517del mouse line shows the hallmarks of CGD and provides a source for Cybb-deficient HSPCs that can be used to evaluate gene-therapy approaches in vitro and in vivo. In a setup using Cas9 RNPs and an AAV repair vector in HSPCs, we show that the mutation can be repaired in 19% of treated cells and that treatment restores ROS production by macrophages. In conclusion, our CybbC517del mouse line provides a new platform for refining and evaluating novel gene therapies and studying X-CGD pathophysiology.


Sujet(s)
Systèmes CRISPR-Cas , Modèles animaux de maladie humaine , Thérapie génétique , Granulomatose septique chronique , NADPH Oxidase 2 , Granulomatose septique chronique/thérapie , Granulomatose septique chronique/génétique , Animaux , Thérapie génétique/méthodes , Souris , NADPH Oxidase 2/génétique , Espèces réactives de l'oxygène/métabolisme , Cellules souches hématopoïétiques/métabolisme , Humains , Macrophages/métabolisme , Mutation
19.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38928171

RÉSUMÉ

Acute myeloid leukemia (AML) is a heterogenous blood cancer with a dismal prognosis. It emanates from leukemic stem cells (LSCs) arising from the genetic transformation of hematopoietic stem cells (HSCs). LSCs hold prognostic value, but their molecular and immunophenotypic heterogeneity poses challenges: there is no single marker for identifying all LSCs across AML samples. We hypothesized that imaging flow cytometry (IFC) paired with artificial intelligence-driven image analysis could visually distinguish LSCs from HSCs based solely on morphology. Initially, a seven-color IFC panel was employed to immunophenotypically identify LSCs and HSCs in bone marrow samples from five AML patients and ten healthy donors, respectively. Next, we developed convolutional neural network (CNN) models for HSC-LSC discrimination using brightfield (BF), side scatter (SSC), and DNA images. Classification using only BF images achieved 86.96% accuracy, indicating significant morphological differences. Accuracy increased to 93.42% when combining BF with DNA images, highlighting differences in nuclear morphology, although DNA images alone were inadequate for accurate HSC-LSC discrimination. Model development using SSC images revealed minor granularity differences. Performance metrics varied substantially between AML patients, indicating considerable morphologic variations among LSCs. Overall, we demonstrate proof-of-concept results for accurate CNN-based HSC-LSC differentiation, instigating the development of a novel technique within AML monitoring.


Sujet(s)
Cytométrie en flux , Cellules souches hématopoïétiques , Leucémie aigüe myéloïde , Cellules souches tumorales , , Humains , Leucémie aigüe myéloïde/anatomopathologie , Cytométrie en flux/méthodes , Cellules souches hématopoïétiques/anatomopathologie , Cellules souches hématopoïétiques/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Immunophénotypage/méthodes , Femelle , Mâle , Traitement d'image par ordinateur/méthodes , Adulte d'âge moyen
20.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38928341

RÉSUMÉ

The purpose of this review is to summarize the current understanding of the therapeutic effect of stem cell-based therapies, including hematopoietic stem cells, for the treatment of ischemic heart damage. Following PRISMA guidelines, we conducted electronic searches in MEDLINE, and EMBASE. We screened 592 studies, and included RCTs, observational studies, and cohort studies that examined the effect of hematopoietic stem cell therapy in adult patients with heart failure. Studies that involved pediatric patients, mesenchymal stem cell therapy, and non-heart failure (HF) studies were excluded from our review. Out of the 592 studies, 7 studies met our inclusion criteria. Overall, administration of hematopoietic stem cells (via intracoronary or myocardial infarct) led to positive cardiac outcomes such as improvements in pathological left-ventricular remodeling, perfusion following acute myocardial infarction, and NYHA symptom class. Additionally, combined death, rehospitalization for heart failure, and infarction were significantly lower in patients treated with bone marrow-derived hematopoietic stem cells. Our review demonstrates that hematopoietic stem cell administration can lead to positive cardiac outcomes for HF patients. Future studies should aim to increase female representation and non-ischemic HF patients.


Sujet(s)
Défaillance cardiaque , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques , Humains , Défaillance cardiaque/thérapie , Défaillance cardiaque/anatomopathologie , Transplantation de cellules souches hématopoïétiques/méthodes , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...