Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cell Rep ; 43(5): 114145, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38669141

RÉSUMÉ

Acute myeloid leukemia (AML) is an aggressive disease with a poor prognosis (5-year survival rate of 30.5% in the United States). Designing cell therapies to target AML is challenging because no single tumor-associated antigen (TAA) is highly expressed on all cancer subpopulations. Furthermore, TAAs are also expressed on healthy cells, leading to toxicity risk. To address these targeting challenges, we engineer natural killer (NK) cells with a multi-input gene circuit consisting of chimeric antigen receptors (CARs) controlled by OR and NOT logic gates. The OR gate kills a range of AML cells from leukemic stem cells to blasts using a bivalent CAR targeting FLT3 and/or CD33. The NOT gate protects healthy hematopoietic stem cells (HSCs) using an inhibitory CAR targeting endomucin, a protective antigen unique to healthy HSCs. NK cells with the combined OR-NOT gene circuit kill multiple AML subtypes and protect primary HSCs, and the circuit also works in vivo.


Sujet(s)
Cellules tueuses naturelles , Leucémie aigüe myéloïde , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Humains , Leucémie aigüe myéloïde/thérapie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/immunologie , Animaux , Souris , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Réseaux de régulation génique , Cellules souches hématopoïétiques/métabolisme , Lignée cellulaire tumorale , Médecine de précision/méthodes , Thérapie cellulaire et tissulaire/méthodes
2.
Mol Cancer Ther ; 20(9): 1508-1520, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34210826

RÉSUMÉ

Advanced peritoneal carcinomatosis including high-grade ovarian cancer has poor prognoses and a poor response rate to current checkpoint inhibitor immunotherapies; thus, there is an unmet need for effective therapeutics that would provide benefit to these patients. Here we present the preclinical development of SENTI-101, a cell preparation of bone marrow-derived mesenchymal stromal (also known as stem) cells (MSC), which are engineered to express two potent immune-modulatory cytokines, IL12 and IL21. Intraperitoneal administration of SENTI-101 results in selective tumor-homing and localized and sustained cytokine production in murine models of peritoneal cancer. SENTI-101 has extended half-life, reduced systemic distribution, and improved antitumor activity when compared with recombinant cytokines, suggesting that it is more effective and has lower risk of systemic immunotoxicities. Treatment of tumor-bearing immune-competent mice with a murine surrogate of SENTI-101 (mSENTI-101) results in a potent and localized immune response consistent with increased number and activation of antigen presenting cells, T cells and B cells, which leads to antitumor response and memory-induced long-term immunity. Consistent with this mechanism of action, co-administration of mSENTI-101 with checkpoint inhibitors leads to synergistic improvement in antitumor response. Collectively, these data warrant potential clinical development of SENTI-101 for patients with peritoneal carcinomatosis and high-grade ovarian cancer.Graphical abstract: SENTI-101 schematic and mechanism of actionSENTI-101 is a novel cell-based immunotherapeutic consisting of bone marrow-derived mesenchymal stromal cells (BM-MSC) engineered to express IL12 and IL21 intended for the treatment of peritoneal carcinomatosis including high-grade serous ovarian cancer. Upon intraperitoneal administration, SENTI-101 homes to peritoneal solid tumors and secretes IL12 and IL21 in a localized and sustained fashion. The expression of these two potent cytokines drives tumor infiltration and engagement of multiple components of the immune system: antigen-presenting cells, T cells, and B cells, resulting in durable antitumor immunity in preclinical models of cancer.


Sujet(s)
Interleukine-12/métabolisme , Interleukines/métabolisme , Mélanome expérimental/immunologie , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/cytologie , Tumeurs/immunologie , Tumeurs du péritoine/immunologie , Animaux , Apoptose , Prolifération cellulaire , Femelle , Humains , Mélanome expérimental/métabolisme , Mélanome expérimental/anatomopathologie , Mélanome expérimental/thérapie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris nude , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/thérapie , Tumeurs du péritoine/métabolisme , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Cell ; 174(1): 156-171.e16, 2018 06 28.
Article de Anglais | MEDLINE | ID: mdl-29909984

RÉSUMÉ

Extracellular proTGF-ß is covalently linked to "milieu" molecules in the matrix or on cell surfaces and is latent until TGF-ß is released by integrins. Here, we show that LRRC33 on the surface of microglia functions as a milieu molecule and enables highly localized, integrin-αVß8-dependent TGF-ß activation. Lrrc33-/- mice lack CNS vascular abnormalities associated with deficiency in TGF-ß-activating integrins but have microglia with a reactive phenotype and after 2 months develop ascending paraparesis with loss of myelinated axons and death by 5 months. Whole bone marrow transplantation results in selective repopulation of Lrrc33-/- brains with WT microglia and halts disease progression. The phenotypes of WT and Lrrc33-/- microglia in the same brain suggest that there is little spreading of TGF-ß activated from one microglial cell to neighboring microglia. Our results suggest that interactions between integrin-bearing cells and cells bearing milieu molecule-associated TGF-ß provide localized and selective activation of TGF-ß.


Sujet(s)
Protéines de transport/métabolisme , Microglie/métabolisme , Système nerveux/métabolisme , Facteur de croissance transformant bêta/métabolisme , Animaux , Axones/métabolisme , Transplantation de moelle osseuse , Encéphale/métabolisme , Protéines de transport/classification , Protéines de transport/génétique , Cellules cultivées , Intégrines/métabolisme , Estimation de Kaplan-Meier , Macrophages/cytologie , Macrophages/immunologie , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Microglie/cytologie , Mutagenèse dirigée , Maladies neurodégénératives/mortalité , Maladies neurodégénératives/anatomopathologie , Maladies neurodégénératives/thérapie , Phylogenèse , Liaison aux protéines , Précurseurs de protéines/génétique , Précurseurs de protéines/métabolisme , Facteur de croissance transformant bêta/génétique
4.
Sci Adv ; 3(12): e1701211, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-29226242

RÉSUMÉ

Recent evidence indicates that hematopoietic stem and progenitor cells (HSPCs) can serve as vehicles for therapeutic molecular delivery to the brain by contributing to the turnover of resident myeloid cell populations. However, such engraftment needs to be fast and efficient to exert its therapeutic potential for diseases affecting the central nervous system. Moreover, the nature of the cells reconstituted after transplantation and whether they could comprise bona fide microglia remain to be assessed. We demonstrate that transplantation of HSPCs in the cerebral lateral ventricles provides rapid engraftment of morphologically, antigenically, and transcriptionally dependable microglia-like cells. We show that the cells comprised within the hematopoietic stem cell compartment and enriched early progenitor fractions generate this microglia-like population when injected in the brain ventricles in the absence of engraftment in the bone marrow. This delivery route has therapeutic relevance because it increases the delivery of therapeutic molecules to the brain, as shown in a humanized animal model of a prototypical lysosomal storage disease affecting the central nervous system.


Sujet(s)
Ventricules cérébraux/cytologie , Transplantation de cellules souches hématopoïétiques/méthodes , Microglie/cytologie , Animaux , Antigènes CD34 , Modèles animaux de maladie humaine , Protéines à fluorescence verte/administration et posologie , Protéines à fluorescence verte/génétique , Cellules souches hématopoïétiques/métabolisme , Humains , Leucodystrophie métachromatique/étiologie , Leucodystrophie métachromatique/thérapie , Souris de lignée C57BL , Souris transgéniques , Cellules myéloïdes/cytologie
5.
Blood ; 130(5): 619-624, 2017 08 03.
Article de Anglais | MEDLINE | ID: mdl-28615219

RÉSUMÉ

The concept that tumor-initiating cells can co-opt the self-renewal program of endogenous stem cells as a means of enforcing their unlimited proliferative potential is widely accepted, yet identification of specific factors that regulate self-renewal of normal and cancer stem cells remains limited. Using a comparative transcriptomic approach, we identify ZNF521/Zfp521 as a conserved hematopoietic stem cell (HSC)-enriched transcription factor in human and murine hematopoiesis whose function in HSC biology remains elusive. Competitive serial transplantation assays using Zfp521-deficient mice revealed that ZFP521 regulates HSC self-renewal and differentiation. In contrast, ectopic expression of ZFP521 in HSCs led to a robust maintenance of progenitor activity in vitro. Transcriptional analysis of human acute myeloid leukemia (AML) patient samples revealed that ZNF521 is highly and specifically upregulated in AMLs with MLL translocations. Using an MLL-AF9 murine leukemia model and serial transplantation studies, we show that ZFP521 is not required for leukemogenesis, although its absence leads to a significant delay in leukemia onset. Furthermore, knockdown of ZNF521 reduced proliferation in human leukemia cell lines possessing MLL-AF9 translocations. Taken together, these results identify ZNF521/ZFP521 as a critical regulator of HSC function, which facilitates MLL-AF9-mediated leukemic disease in mice.


Sujet(s)
Prolifération cellulaire , Protéines de liaison à l'ADN/métabolisme , Cellules souches hématopoïétiques/métabolisme , Tumeurs expérimentales/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Facteurs de transcription/métabolisme , Animaux , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Cellules souches hématopoïétiques/anatomopathologie , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme , Humains , Souris , Souris knockout , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Tumeurs expérimentales/anatomopathologie , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Facteurs de transcription/génétique , Translocation génétique
6.
Cell Stem Cell ; 15(5): 643-52, 2014 Nov 06.
Article de Anglais | MEDLINE | ID: mdl-25517468

RÉSUMÉ

Genome editing via CRISPR/Cas9 has rapidly become the tool of choice by virtue of its efficacy and ease of use. However, CRISPR/Cas9-mediated genome editing in clinically relevant human somatic cells remains untested. Here, we report CRISPR/Cas9 targeting of two clinically relevant genes, B2M and CCR5, in primary human CD4+ T cells and CD34+ hematopoietic stem and progenitor cells (HSPCs). Use of single RNA guides led to highly efficient mutagenesis in HSPCs but not in T cells. A dual guide approach improved gene deletion efficacy in both cell types. HSPCs that had undergone genome editing with CRISPR/Cas9 retained multilineage potential. We examined predicted on- and off-target mutations via target capture sequencing in HSPCs and observed low levels of off-target mutagenesis at only one site. These results demonstrate that CRISPR/Cas9 can efficiently ablate genes in HSPCs with minimal off-target mutagenesis, which could have broad applicability for hematopoietic cell-based therapy.


Sujet(s)
Protéines associées aux CRISPR/métabolisme , Clustered regularly interspaced short palindromic repeats/génétique , Délétion de gène , Cellules souches hématopoïétiques/métabolisme , Animaux , Antigènes CD34/métabolisme , Lignage cellulaire , Cellules cultivées , Ciblage de gène , Locus génétiques , Génome humain/génétique , Cellules souches hématopoïétiques/cytologie , Séquençage nucléotidique à haut débit , Humains , Souris , Édition des ARN/génétique , /métabolisme , Récepteurs CCR5/métabolisme
7.
J Clin Invest ; 124(7): 2846-8, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24937422

RÉSUMÉ

The regenerative capacity of tissues to recover from injury or stress is dependent on stem cell competence, yet the underlying mechanisms that govern how stem cells detect stress and initiate appropriate responses are poorly understood. In this issue of the JCI, Cho and Yusuf et al. demonstrate that the purinergic receptor P2Y14 may mediate the hematopoietic stem and progenitor cell regenerative response.


Sujet(s)
Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/effets des radiations , Récepteurs purinergiques P2Y/métabolisme , Animaux , Femelle , Mâle , Grossesse
8.
Cell ; 157(3): 549-64, 2014 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-24766805

RÉSUMÉ

Hematopoietic stem cells (HSCs) sustain blood formation throughout life and are the functional units of bone marrow transplantation. We show that transient expression of six transcription factors Run1t1, Hlf, Lmo2, Prdm5, Pbx1, and Zfp37 imparts multilineage transplantation potential onto otherwise committed lymphoid and myeloid progenitors and myeloid effector cells. Inclusion of Mycn and Meis1 and use of polycistronic viruses increase reprogramming efficacy. The reprogrammed cells, designated induced-HSCs (iHSCs), possess clonal multilineage differentiation potential, reconstitute stem/progenitor compartments, and are serially transplantable. Single-cell analysis revealed that iHSCs derived under optimal conditions exhibit a gene expression profile that is highly similar to endogenous HSCs. These findings demonstrate that expression of a set of defined factors is sufficient to activate the gene networks governing HSC functional identity in committed blood cells. Our results raise the prospect that blood cell reprogramming may be a strategy for derivation of transplantable stem cells for clinical application.


Sujet(s)
Reprogrammation cellulaire , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Facteurs de transcription/métabolisme , Animaux , Transplantation de cellules souches hématopoïétiques , Protéines à homéodomaine/génétique , Souris , Souris de lignée C57BL , Protéine du site-1 d'intégration des virus myéloïdes écotropiques , Protéine du proto-oncogène N-Myc , Protéines tumorales/génétique , Protéines proto-oncogènes/génétique , Analyse sur cellule unique , Transcriptome
9.
Stem Cell Reports ; 1(3): 266-80, 2013.
Article de Anglais | MEDLINE | ID: mdl-24319662

RÉSUMÉ

Hematopoietic stem cells (HSCs) maintain blood homeostasis and are the functional units of bone marrow transplantation. To improve the molecular understanding of HSCs and their proximal progenitors, we performed transcriptome analysis within the context of the ImmGen Consortium data set. Gene sets that define steady-state and mobilized HSCs, as well as hematopoietic stem and progenitor cells (HSPCs), were determined. Genes involved in transcriptional regulation, including a group of putative transcriptional repressors, were identified in multipotent progenitors and HSCs. Proximal promoter analyses combined with ImmGen module analysis identified candidate regulators of HSCs. Enforced expression of one predicted regulator, Hlf, in diverse HSPC subsets led to extensive self-renewal activity ex vivo. These analyses reveal unique insights into the mechanisms that control the core properties of HSPCs.


Sujet(s)
Analyse de profil d'expression de gènes , Régulation de l'expression des gènes au cours du développement/génétique , Cellules souches hématopoïétiques , Cellules souches , Transplantation de moelle osseuse , Différenciation cellulaire , Lignage cellulaire , Régions promotrices (génétique)
10.
Cell Stem Cell ; 12(4): 413-25, 2013 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-23415915

RÉSUMÉ

The functional potential of hematopoietic stem cells (HSCs) declines during aging, and in doing so, significantly contributes to hematopoietic pathophysiology in the elderly. To explore the relationship between age-associated HSC decline and the epigenome, we examined global DNA methylation of HSCs during ontogeny in combination with functional analysis. Although the DNA methylome is generally stable during aging, site-specific alterations of DNA methylation occur at genomic regions associated with hematopoietic lineage potential and selectively target genes expressed in downstream progenitor and effector cells. We found that age-associated HSC decline, replicative limits, and DNA methylation are largely dependent on the proliferative history of HSCs, yet appear to be telomere-length independent. Physiological aging and experimentally enforced proliferation of HSCs both led to DNA hypermethylation of genes regulated by Polycomb Repressive Complex 2. Our results provide evidence that epigenomic alterations of the DNA methylation landscape contribute to the functional decline of HSCs during aging.


Sujet(s)
Vieillissement de la cellule/génétique , Méthylation de l'ADN/génétique , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Animaux , Prolifération cellulaire , Réplication de l'ADN/génétique , Régulation de l'expression des gènes au cours du développement , Hématopoïèse/génétique , Mâle , Souris , Souris de lignée C57BL , Modèles biologiques , Complexe répresseur Polycomb-2/métabolisme , Télomère/métabolisme
11.
Cell Metab ; 15(1): 2-3, 2012 Jan 04.
Article de Anglais | MEDLINE | ID: mdl-22225868

RÉSUMÉ

While age-dependent stem cell decline is widely recognized as being a key component of organismal aging, the underlying mechanisms remain elusive. In this issue of Cell Metabolism, Suomalainen and colleagues provide evidence that mitochondrial mutation and associated reactive oxygen species can adversely impact tissue-specific stem and progenitor cell function.

13.
Mol Cell Biol ; 27(24): 8824-33, 2007 Dec.
Article de Anglais | MEDLINE | ID: mdl-17938204

RÉSUMÉ

The Sleeping Beauty (SB) transposon represents an important vehicle for in vivo gene delivery because it can efficiently and stably integrate into mammalian genomes. In this report, we examined transposon expression in human cells using a novel nonselective fluorescence-activated cell sorter-based method and discovered that SB integrates approximately 20 times more frequently than previously reported within systems that were dependent on transgene expression and likely subject to postintegrative gene silencing. Over time, phenotypic analysis of clonal integrants demonstrated that SB undergoes additional postintegrative gene silencing, which varied based on the promoter used for transgene expression. Molecular and biochemical studies suggested that transposon silencing was influenced by DNA methylation and histone deacetylation because both 5-aza-2'-deoxycytidine and trichostatin A partially rescued transgene silencing in clonal cell lines. Collectively, these data reveal the existence of a multicomponent postintegrative gene silencing network that efficiently targets invading transposon sequences for transcriptional silencing in mammalian cells.


Sujet(s)
Éléments transposables d'ADN/génétique , Extinction de l'expression des gènes , Recombinaison génétique , Transposases/génétique , Acétylation , Animaux , Méthylation de l'ADN , Analyse de profil d'expression de gènes , Cellules HeLa , Histone/métabolisme , Humains , Mammifères , Mutagenèse par insertion , Transgènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE