Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Ther ; 32(6): 1628-1642, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38556793

ABSTRACT

Severe congenital neutropenia (CN) is an inherited pre-leukemia bone marrow failure syndrome commonly caused by autosomal-dominant ELANE mutations (ELANE-CN). ELANE-CN patients are treated with daily injections of recombinant human granulocyte colony-stimulating factor (rhG-CSF). However, some patients do not respond to rhG-CSF, and approximately 15% of ELANE-CN patients develop myelodysplasia or acute myeloid leukemia. Here, we report the development of a curative therapy for ELANE-CN through inhibition of ELANE mRNA expression by introducing two single-strand DNA breaks at the opposing DNA strands of the ELANE promoter TATA box using CRISPR-Cas9D10A nickases-termed MILESTONE. This editing effectively restored defective neutrophil differentiation of ELANE-CN CD34+ hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, without affecting the functions of the edited neutrophils. CRISPResso analysis of the edited ELANE-CN CD34+ HSPCs revealed on-target efficiencies of over 90%. Simultaneously, GUIDE-seq, CAST-Seq, and rhAmpSeq indicated a safe off-target profile with no off-target sites or chromosomal translocations. Taken together, ex vivo gene editing of ELANE-CN HSPCs using MILESTONE in the setting of autologous stem cell transplantation could be a universal, safe, and efficient gene therapy approach for ELANE-CN patients.


Subject(s)
CRISPR-Cas Systems , Congenital Bone Marrow Failure Syndromes , Gene Editing , Genetic Therapy , Leukocyte Elastase , Neutropenia , Promoter Regions, Genetic , Gene Editing/methods , Humans , Neutropenia/congenital , Neutropenia/therapy , Neutropenia/genetics , Genetic Therapy/methods , Congenital Bone Marrow Failure Syndromes/therapy , Congenital Bone Marrow Failure Syndromes/genetics , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Animals , Mice , Neutrophils/metabolism , Hematopoietic Stem Cells/metabolism , Mutation , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/genetics , Genetic Diseases, X-Linked/therapy , Genetic Diseases, X-Linked/genetics
5.
Nat Commun ; 13(1): 2948, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35618709

ABSTRACT

Protein therapeutics frequently face major challenges, including complicated production, instability, poor solubility, and aggregation. De novo protein design can readily address these challenges. Here, we demonstrate the utility of a topological refactoring strategy to design novel granulopoietic proteins starting from the granulocyte-colony stimulating factor (G-CSF) structure. We change a protein fold by rearranging the sequence and optimising it towards the new fold. Testing four designs, we obtain two that possess nanomolar activity, the most active of which is highly thermostable and protease-resistant, and matches its designed structure to atomic accuracy. While the designs possess starkly different sequence and structure from the native G-CSF, they show specific activity in differentiating primary human haematopoietic stem cells into mature neutrophils. The designs also show significant and specific activity in vivo. Our topological refactoring approach is largely independent of sequence or structural context, and is therefore applicable to a wide range of protein targets.


Subject(s)
Granulocyte Colony-Stimulating Factor , Hematopoiesis , Granulocyte Colony-Stimulating Factor/genetics , Hematopoietic Stem Cells , Humans , Neutrophils
6.
Cell Stem Cell ; 28(5): 906-922.e6, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33894142

ABSTRACT

Severe congenital neutropenia (CN) is a pre-leukemic bone marrow failure syndrome that can evolve to acute myeloid leukemia (AML). Mutations in CSF3R and RUNX1 are frequently observed in CN patients, although how they drive the transition from CN to AML (CN/AML) is unclear. Here we establish a model of stepwise leukemogenesis in CN/AML using CRISPR-Cas9 gene editing of CN patient-derived iPSCs. We identified BAALC upregulation and resultant phosphorylation of MK2a as a key leukemogenic event. BAALC deletion or treatment with CMPD1, a selective inhibitor of MK2a phosphorylation, blocked proliferation and induced differentiation of primary CN/AML blasts and CN/AML iPSC-derived hematopoietic stem and progenitor cells (HSPCs) without affecting healthy donor or CN iPSC-derived HSPCs. Beyond detailing a useful method for future investigation of stepwise leukemogenesis, this study suggests that targeting BAALC and/or MK2a phosphorylation may prevent leukemogenic transformation or eliminate AML blasts in CN/AML and RUNX1 mutant BAALC(hi) de novo AML.


Subject(s)
Induced Pluripotent Stem Cells , Leukemia, Myeloid, Acute , Neoplasm Proteins , Neutropenia , Congenital Bone Marrow Failure Syndromes , Humans , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Neoplasm Proteins/genetics , Neutropenia/congenital , Neutropenia/genetics , Oncogenes
7.
Stem Cell Res Ther ; 12(1): 112, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33546767

ABSTRACT

BACKGROUND: Nicotinamide phosphoribosyltransferase (NAMPT) regulates cellular functions through the protein deacetylation activity of nicotinamide adenine dinucleotide (NAD+)-dependent sirtuins (SIRTs). SIRTs regulate functions of histones and none-histone proteins. The role of NAMPT/SIRT pathway in the regulation of maintenance and differentiation of human-induced pluripotent stem (iPS) cells is not fully elucidated. METHODS: We evaluated the effects of specific inhibitors of NAMPT or SIRT2 on the pluripotency, proliferation, survival, and hematopoietic differentiation of human iPS cells. We also studied the molecular mechanism downstream of NAMPT/SIRTs in iPS cells. RESULTS: We demonstrated that NAMPT is indispensable for the maintenance, survival, and hematopoietic differentiation of iPS cells. We found that inhibition of NAMPT or SIRT2 in iPS cells induces p53 protein by promoting its lysine acetylation. This leads to activation of the p53 target, p21, with subsequent cell cycle arrest and induction of apoptosis in iPS cells. NAMPT and SIRT2 inhibition also affect hematopoietic differentiation of iPS cells in an embryoid body (EB)-based cell culture system. CONCLUSIONS: Our data demonstrate the essential role of the NAMPT/SIRT2/p53/p21 signaling axis in the maintenance and hematopoietic differentiation of iPS cells.


Subject(s)
Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells , Cell Differentiation , Cytokines/genetics , Cytokines/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Signal Transduction , Sirtuin 2/genetics , Sirtuin 2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
PLoS Biol ; 18(12): e3000919, 2020 12.
Article in English | MEDLINE | ID: mdl-33351791

ABSTRACT

Computational protein design is rapidly becoming more powerful, and improving the accuracy of computational methods would greatly streamline protein engineering by eliminating the need for empirical optimization in the laboratory. In this work, we set out to design novel granulopoietic agents using a rescaffolding strategy with the goal of achieving simpler and more stable proteins. All of the 4 experimentally tested designs were folded, monomeric, and stable, while the 2 determined structures agreed with the design models within less than 2.5 Å. Despite the lack of significant topological or sequence similarity to their natural granulopoietic counterpart, 2 designs bound to the granulocyte colony-stimulating factor (G-CSF) receptor and exhibited potent, but delayed, in vitro proliferative activity in a G-CSF-dependent cell line. Interestingly, the designs also induced proliferation and differentiation of primary human hematopoietic stem cells into mature granulocytes, highlighting the utility of our approach to develop highly active therapeutic leads purely based on computational design.


Subject(s)
Granulocytes/cytology , Protein Engineering/methods , Cell Differentiation , Cells, Cultured , Computational Biology/methods , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/drug effects , Hematopoiesis/drug effects , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Humans , Neutrophils , Structure-Activity Relationship
9.
Ann N Y Acad Sci ; 1466(1): 83-92, 2020 04.
Article in English | MEDLINE | ID: mdl-32083314

ABSTRACT

Cyclic neutropenia (CyN) is a hematologic disorder in which peripheral blood absolute neutrophil counts (ANCs) show cycles of approximately 21-day intervals. The majority of CyN patients harbor ELANE mutations, but the mechanism of ANC cycling is unclear. We performed analysis of bone marrow (BM) subpopulations in CyN patients at the peak and the nadir of the ANC cycle and detected high proportions of BM hematopoietic stem cells (HSCs) and hematopoietic stem and progenitor cells (HSPCs) at the nadir of the ANC cycle, as compared with the peak. BM HSPCs produced fewer granulocyte colony-forming unit colonies at the ANC peak. To investigate the mechanism of cycling, we found that mRNA expression levels of ELANE and unfolded protein response (UPR)-related genes (ATF6, BiP (HSPA5), CHOP (DDIT3), and PERK (EIF2AK3)) were elevated, but antiapoptotic genes (Bcl-2 (BCL2) and bcl-xL (BCL2L1)) were reduced in CD34+ cells tested at the ANC nadir. Moreover, HSPCs revealed increased levels of reactive oxygen species and gH2AX at the ANC nadir. We suggest that in CyN patients, some HSPCs escape the UPR-induced endoplasmic reticulum (ER) stress and proliferate in response to granulocyte colony-stimulating factor (G-CSF) to a certain threshold at which UPR again affects the majority of HSPCs. There is a cyclic balance between ER stress-induced apoptosis of HSPCs and compensatory G-CSF-stimulated HSPC proliferation followed by granulocytic differentiation.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Leukocyte Elastase/genetics , Neutropenia/etiology , Unfolded Protein Response/physiology , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Follow-Up Studies , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Humans , Leukocyte Elastase/physiology , Mutation , Neutropenia/drug therapy , Neutropenia/metabolism , Neutropenia/pathology , Reactive Oxygen Species/metabolism , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/physiology , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
10.
Methods Mol Biol ; 2115: 455-469, 2020.
Article in English | MEDLINE | ID: mdl-32006417

ABSTRACT

In this chapter, we present an optimized CRISPR/Cas9 RNP nucleofection approach for gene knockout (KO) in hematopoietic stem and progenitor cells (HSPCs). With experimentally proved active locus-specific sgRNAs, we routinely reach over 80% gene KO in HSPCs, thus avoiding the need for cell sorting or enrichment of targeted cell population. Additionally, we provide a protocol for in vitro granulocytic differentiation of HSPCs after gene KO and detailed description of granulocyte function tests which can be applied to study the effects of a particular gene KO.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Leukopoiesis , Cells, Cultured , Gene Knockout Techniques/methods , Granulocytes/metabolism , Hematopoietic Stem Cells/metabolism , Humans , RNA, Guide, Kinetoplastida/genetics
11.
Haematologica ; 105(3): 598-609, 2020 03.
Article in English | MEDLINE | ID: mdl-31248972

ABSTRACT

A Autosomal-dominant ELANE mutations are the most common cause of severe congenital neutropenia. Although the majority of congenital neutropenia patients respond to daily granulocyte colony stimulating factor, approximately 15 % do not respond to this cytokine at doses up to 50 µg/kg/day and approximately 15 % of patients will develop myelodysplasia or acute myeloid leukemia. "Maturation arrest," the failure of the marrow myeloid progenitors to form mature neutrophils, is a consistent feature of ELANE associated congenital neutropenia. As mutant neutrophil elastase is the cause of this abnormality, we hypothesized that ELANE associated neutropenia could be treated and "maturation arrest" corrected by a CRISPR/Cas9-sgRNA ribonucleoprotein mediated ELANE knockout. To examine this hypothesis, we used induced pluripotent stem cells from two congenital neutropenia patients and primary hematopoietic stem and progenitor cells from four congenital neutropenia patients harboring ELANE mutations as well as HL60 cells expressing mutant ELANE We observed that granulocytic differentiation of ELANE knockout induced pluripotent stem cells and primary hematopoietic stem and progenitor cells were comparable to healthy individuals. Phagocytic functions, ROS production, and chemotaxis of the ELANE KO (knockout) neutrophils were also normal. Knockdown of ELANE in the mutant ELANE expressing HL60 cells also allowed full maturation and formation of abundant neutrophils. These observations suggest that ex vivo CRISPR/Cas9 RNP based ELANE knockout of patients' primary hematopoietic stem and progenitor cells followed by autologous transplantation may be an alternative therapy for congenital neutropenia.


Subject(s)
Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells , Neutropenia , CRISPR-Cas Systems , Congenital Bone Marrow Failure Syndromes , Humans , Mutation , Neutropenia/congenital , Neutropenia/genetics
12.
Blood Adv ; 3(1): 63-71, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622144

ABSTRACT

CRISPR/Cas9-mediated gene editing of stem cells and primary cell types has several limitations for clinical applications. The direct delivery of ribonucleoprotein (RNP) complexes consisting of Cas9 nuclease and guide RNA (gRNA) has improved DNA- and virus-free gene modifications, but it does not enable the essential enrichment of the gene-edited cells. Here, we established a protocol for the fluorescent labeling and delivery of CRISPR/Cas9-gRNA RNP in primary human hematopoietic stem and progenitor cells (HSPCs) and induced pluripotent stem cells (iPSCs). As a proof of principle for genes with low-abundance transcripts and context-dependent inducible expression, we successfully deleted growth arrest and DNA-damage-inducible ß (GADD45B). We found that GADD45B is indispensable for DNA damage protection and survival in stem cells. Thus, we describe an easy and efficient protocol of DNA-free gene editing of hard-to-target transcripts and enrichment of gene-modified cells that are generally difficult to transfect.


Subject(s)
Antigens, Differentiation/genetics , CRISPR-Cas Systems , Gene Knockout Techniques , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Ribonucleoproteins/metabolism , Stress, Physiological/genetics , Antigens, Differentiation/metabolism , Cell Line , DNA Damage , Gene Editing/methods , Gene Targeting/methods , Humans , Macromolecular Substances/metabolism , Protein Binding , RNA, Guide, Kinetoplastida/genetics , Stress, Physiological/radiation effects
13.
Exp Hematol ; 71: 51-60, 2019 03.
Article in English | MEDLINE | ID: mdl-30615903

ABSTRACT

We describe the establishment of an embryoid-body-based protocol for hematopoietic/myeloid differentiation of human induced pluripotent stem cells that allows the generation of CD34+ cells or mature myeloid cells in vitro. Using this model, we were able to recapitulate the defective granulocytic differentiation in patients with severe congenital neutropenia (CN), an inherited preleukemia bone marrow failure syndrome. Importantly, in vitro maturation arrest of granulopoiesis was associated with an elevated unfolded protein response (UPR) and enhanced expression of the cell cycle inhibitor p21. Consistent with this, we found that CD34+ cells of CN patients were highly susceptible to DNA damage and showed diminished DNA repair. These observations suggest that targeting the UPR pathway or inhibiting DNA damage might protect hematopoietic cells of CN patients from leukemogenic transformation, at least to some extent.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA Damage , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Leukemia/etiology , Models, Biological , Neutropenia/congenital , Unfolded Protein Response , Antigens, CD34/metabolism , Biomarkers , Cells, Cultured , Cellular Reprogramming , Congenital Bone Marrow Failure Syndromes , Endoplasmic Reticulum Stress , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/pathology , Leukemia/metabolism , Leukemia/pathology , Neutropenia/etiology , Neutropenia/metabolism , Neutropenia/pathology
14.
Ann Hematol ; 96(3): 345-353, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27966038

ABSTRACT

Severe congenital neutropenia (CN) is a bone marrow failure syndrome characterized by an absolute neutrophil count (ANC) below 500 cells/µL and recurrent, life-threatening bacterial infections. Treatment with granulocyte colony-stimulating factor (G-CSF) increases the ANC in the majority of CN patients. In contrary, granulocyte-monocyte colony-stimulating factor (GM-CSF) fails to increase neutrophil numbers in CN patients in vitro and in vivo, suggesting specific defects in signaling pathways downstream of GM-CSF receptor. Recently, we detected that G-CSF induces granulopoiesis in CN patients by hyperactivation of nicotinamide phosphoribosyl transferase (NAMPT)/Sirtuin 1 signaling in myeloid cells. Here, we demonstrated that, in contrast to G-CSF, GM-CSF failed to induce NAMPT-dependent granulopoiesis in CN patients. We further identified NAMPT signaling as an essential downstream effector of the GM-CSF pathway in myelopoiesis.


Subject(s)
Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neutropenia/congenital , Nicotinamide Phosphoribosyltransferase/metabolism , Signal Transduction/physiology , Cells, Cultured , Congenital Bone Marrow Failure Syndromes , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neutropenia/drug therapy , Neutropenia/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Signal Transduction/drug effects , Treatment Outcome
15.
Oncotarget ; 6(41): 43540-56, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26498353

ABSTRACT

The transcription factor SOX2 is a key regulator of pluripotency in embryonic stem cells and plays important roles in early organogenesis. Recently, SOX2 expression was documented in various cancers and suggested as a cancer stem cell (CSC) marker. Here we identify the Ser/Thr-kinase AKT as an upstream regulator of SOX2 protein turnover in breast carcinoma (BC). SOX2 and pAKT are co-expressed and co-regulated in breast CSCs and depletion of either reduces clonogenicity. Ectopic SOX2 expression restores clonogenicity and in vivo tumorigenicity of AKT-inhibited cells, suggesting that SOX2 acts as a functional downstream AKT target. Mechanistically, we show that AKT physically interacts with the SOX2 protein to modulate its subcellular distribution. AKT kinase inhibition results in enhanced cytoplasmic retention of SOX2, presumably via impaired nuclear import, and in successive cytoplasmic proteasomal degradation of the protein. In line, blockade of either nuclear transport or proteasomal degradation rescues SOX2 expression in AKT-inhibited BC cells. Finally, AKT inhibitors efficiently suppress the growth of SOX2-expressing putative cancer stem cells, whereas conventional chemotherapeutics select for this population. Together, our results suggest the AKT/SOX2 molecular axis as a regulator of BC clonogenicity and AKT inhibitors as promising drugs for the treatment of SOX2-positive BC.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/physiology , Heterografts , Humans , Immunoblotting , Immunoprecipitation , Neoplastic Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Transduction, Genetic , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...