Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 491
Filter
1.
Commun Biol ; 7(1): 525, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702433

ABSTRACT

Disabled 2 (Dab2), an adaptor protein, is up regulated in the hair follicle stem cells (HFSCs); however, its role in any tissue stem cells has not been studied. In the present study, we have reported that Dab2 conditional knockout (Dab2-cKO) mice exhibited a delay in the HF cycle due to perturbed activation of HFSCs. Further, Dab2-cKO mice showed a reduction in the number of HFSCs and reduced colony forming ability of HFSCs. Dab2-cKO mice showed extended quiescence of HFSCs concomitant with an increased expression of Nfatc1. Dab2-cKO mice showed a decreased expression of anti-aging genes such as Col17a1, decorin, Sirt2 and Sirt7. Dab2-cKO mice did not show full hair coat recovery in aged mice thereby suggesting an accelerated aging process. Overall, we unveil for the first time, the role of Dab2 that regulate activation and self-renewal of HFSCs.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Hair Follicle , Mice, Knockout , Stem Cells , Animals , Hair Follicle/metabolism , Hair Follicle/cytology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Stem Cells/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Cell Self Renewal/genetics , Mice, Inbred C57BL , Cell Proliferation
2.
Cell Stem Cell ; 31(5): 754-771.e6, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701759

ABSTRACT

Development of embryonic stem cells (ESCs) into neurons requires intricate regulation of transcription, splicing, and translation, but how these processes interconnect is not understood. We found that polypyrimidine tract binding protein 1 (PTBP1) controls splicing of DPF2, a subunit of BRG1/BRM-associated factor (BAF) chromatin remodeling complexes. Dpf2 exon 7 splicing is inhibited by PTBP1 to produce the DPF2-S isoform early in development. During neuronal differentiation, loss of PTBP1 allows exon 7 inclusion and DPF2-L expression. Different cellular phenotypes and gene expression programs were induced by these alternative DPF2 isoforms. We identified chromatin binding sites enriched for each DPF2 isoform, as well as sites bound by both. In ESC, DPF2-S preferential sites were bound by pluripotency factors. In neuronal progenitors, DPF2-S sites were bound by nuclear factor I (NFI), while DPF2-L sites were bound by CCCTC-binding factor (CTCF). DPF2-S sites exhibited enhancer modifications, while DPF2-L sites showed promoter modifications. Thus, alternative splicing redirects BAF complex targeting to impact chromatin organization during neuronal development.


Subject(s)
Alternative Splicing , Cell Differentiation , Chromatin , Heterogeneous-Nuclear Ribonucleoproteins , Neurons , Polypyrimidine Tract-Binding Protein , Transcription Factors , Alternative Splicing/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Mice , Neurons/metabolism , Neurons/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription, Genetic , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Exons/genetics , Humans , Cell Self Renewal/genetics
3.
Nat Commun ; 15(1): 3432, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653778

ABSTRACT

Temporal regulation of super-enhancer (SE) driven transcription factors (TFs) underlies normal developmental programs. Neuroblastoma (NB) arises from an inability of sympathoadrenal progenitors to exit a self-renewal program and terminally differentiate. To identify SEs driving TF regulators, we use all-trans retinoic acid (ATRA) to induce NB growth arrest and differentiation. Time-course H3K27ac ChIP-seq and RNA-seq reveal ATRA coordinated SE waves. SEs that decrease with ATRA link to stem cell development (MYCN, GATA3, SOX11). CRISPR-Cas9 and siRNA verify SOX11 dependency, in vitro and in vivo. Silencing the SOX11 SE using dCAS9-KRAB decreases SOX11 mRNA and inhibits cell growth. Other TFs activate in sequential waves at 2, 4 and 8 days of ATRA treatment that regulate neural development (GATA2 and SOX4). Silencing the gained SOX4 SE using dCAS9-KRAB decreases SOX4 expression and attenuates ATRA-induced differentiation genes. Our study identifies oncogenic lineage drivers of NB self-renewal and TFs critical for implementing a differentiation program.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Neoplastic , Neuroblastoma , SOXC Transcription Factors , Tretinoin , Neuroblastoma/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Tretinoin/pharmacology , Tretinoin/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Humans , Animals , Cell Line, Tumor , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/genetics , Cell Lineage/genetics , GATA2 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , CRISPR-Cas Systems , N-Myc Proto-Oncogene Protein/metabolism , N-Myc Proto-Oncogene Protein/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics
4.
Nucleic Acids Res ; 52(7): 3667-3681, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38321961

ABSTRACT

The Wnt/ß-Catenin pathway plays a key role in cell fate determination during development and in adult tissue regeneration by stem cells. These processes involve profound gene expression and epigenome remodeling and linking Wnt/ß-Catenin signaling to chromatin modifications has been a challenge over the past decades. Functional studies of the lysine demethylase LSD1/KDM1A converge to indicate that this epigenetic regulator is a key regulator of cell fate, although the extracellular cues controlling LSD1 action remain largely unknown. Here we show that ß-Catenin is a substrate of LSD1. Demethylation by LSD1 prevents ß-Catenin degradation thereby maintaining its nuclear levels. Consistently, in absence of LSD1, ß-Catenin transcriptional activity is reduced in both MuSCs and ESCs. Moreover, inactivation of LSD1 in mouse muscle stem cells and embryonic stem cells shows that LSD1 promotes mitotic spindle orientation via ß-Catenin protein stabilization. Altogether, by inscribing LSD1 and ß-Catenin in the same molecular cascade linking extracellular factors to gene expression, our results provide a mechanistic explanation to the similarity of action of canonical Wnt/ß-Catenin signaling and LSD1 on stem cell fate.


Subject(s)
Cell Self Renewal , Histone Demethylases , Wnt Signaling Pathway , beta Catenin , Animals , Histone Demethylases/metabolism , Histone Demethylases/genetics , beta Catenin/metabolism , beta Catenin/genetics , Mice , Cell Self Renewal/genetics , Cell Nucleus/metabolism , Spindle Apparatus/metabolism , Cell Differentiation/genetics , Humans , Stem Cells/metabolism , Stem Cells/cytology
5.
J Hematol Oncol ; 17(1): 7, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38302992

ABSTRACT

BACKGROUND: While liver cancer stem cells (CSCs) play a crucial role in hepatocellular carcinoma (HCC) initiation, progression, recurrence, and treatment resistance, the mechanism underlying liver CSC self-renewal remains elusive. We aim to characterize the role of Methyltransferase 16 (METTL16), a recently identified RNA N6-methyladenosine (m6A) methyltransferase, in HCC development/maintenance, CSC stemness, as well as normal hepatogenesis. METHODS: Liver-specific Mettl16 conditional KO (cKO) mice were generated to assess its role in HCC pathogenesis and normal hepatogenesis. Hydrodynamic tail-vein injection (HDTVi)-induced de novo hepatocarcinogenesis and xenograft models were utilized to determine the role of METTL16 in HCC initiation and progression. A limiting dilution assay was utilized to evaluate CSC frequency. Functionally essential targets were revealed via integrative analysis of multi-omics data, including RNA-seq, RNA immunoprecipitation (RIP)-seq, and ribosome profiling. RESULTS: METTL16 is highly expressed in liver CSCs and its depletion dramatically decreased CSC frequency in vitro and in vivo. Mettl16 KO significantly attenuated HCC initiation and progression, yet only slightly influenced normal hepatogenesis. Mechanistic studies, including high-throughput sequencing, unveiled METTL16 as a key regulator of ribosomal RNA (rRNA) maturation and mRNA translation and identified eukaryotic translation initiation factor 3 subunit a (eIF3a) transcript as a bona-fide target of METTL16 in HCC. In addition, the functionally essential regions of METTL16 were revealed by CRISPR gene tiling scan, which will pave the way for the development of potential inhibitor(s). CONCLUSIONS: Our findings highlight the crucial oncogenic role of METTL16 in promoting HCC pathogenesis and enhancing liver CSC self-renewal through augmenting mRNA translation efficiency.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Neoplastic Stem Cells , Animals , Humans , Mice , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Self Renewal/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Methyltransferases/genetics , Methyltransferases/metabolism , Neoplastic Stem Cells/pathology , Protein Biosynthesis , Ribosomes/metabolism , RNA
7.
Haematologica ; 109(2): 411-421, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37584287

ABSTRACT

Leukemia stem cells (LSC) represent a crucial and rare subset of cells present in acute myeloid leukemia (AML); they play a pivotal role in the initiation, maintenance, and relapse of this disease. Targeting LSC holds great promise for preventing AML relapse and improving long-term outcomes. However the precise molecular mechanisms governing LSC self-renewal are still poorly understood. Here, we present compelling evidence that the expression of miR-30e-5p, a potential tumor-suppressive microRNA, is significantly lower in AML samples than in healthy bone marrow samples. Forced expression of miR- 30e effectively inhibits leukemogenesis, impairs LSC self-renewal, and delays leukemia progression. Mechanistically, Cyb561 acts as a direct target of miR-30e-5p in LSC, and its deficiency restricts the self-renewal of LSC by activating reactive oxygen series signaling and markedly prolongs recipients' survival. Moreover, genetic or pharmacological overexpression of miR-30e-5p or knockdown of Cyb561 suppresses the growth of human AML cells. In conclusion, our findings establish the crucial role of the miR-30e-5p/Cyb561/ROS axis in finely regulating LSC self-renewal, highlighting Cyb561 as a potential therapeutic target for LSC-directed therapies.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , Humans , Reactive Oxygen Species , Cell Self Renewal/genetics , MicroRNAs/genetics , Signal Transduction , Recurrence , Cell Proliferation/genetics , Cell Line, Tumor
8.
Cancer Biol Ther ; 24(1): 2271638, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37927213

ABSTRACT

The poly(rC) binding protein 1 gene (PCBP1) encodes the heterogeneous nuclear ribonucleoprotein E1 (hnRNPE1), a nucleic acid-binding protein that plays a tumor-suppressive role in the mammary epithelium by regulating phenotypic plasticity and cell fate. Following the loss of PCBP1 function, the FAM3C gene (encoding the Interleukin-like EMT inducer, or "ILEI" protein) and the leukemia inhibitory factor receptor (LIFR) gene are upregulated. Interaction between FAM3C and LIFR in the extracellular space induces phosphorylation of signal transducer and activator of transcription 3 (pSTAT3). Overexpression and/or hyperactivity of STAT3 has been detected in 40% of breast cancer cases and is associated with a poor prognosis. Herein, we characterize feed-forward regulation of LIFR expression in response to FAM3C/LIFR/STAT3 signaling in mammary epithelial cells. We show that PCBP1 upregulates LIFR transcription through activity at the LIFR promoter, and that FAM3C participates in transcriptional regulation of LIFR. Additionally, our bioinformatic analysis reveals a signature of transcriptional regulation associated with FAM3C/LIFR interaction and identifies the TWIST1 transcription factor as a downstream effector that participates in the maintenance of LIFR expression. Finally, we characterize the effect of LIFR expression in cell-based experiments that demonstrate the promotion of invasion, migration, and self-renewal of breast cancer stem cells (BCSCs), consistent with previous studies linking LIFR expression to tumor initiation and metastasis in mammary epithelial cells.


Subject(s)
Breast Neoplasms , DNA-Binding Proteins , RNA-Binding Proteins , Female , Humans , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Self Renewal/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Neoplasm Proteins/genetics , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Neoplasm Invasiveness
9.
Mol Med Rep ; 27(4)2023 Apr.
Article in English | MEDLINE | ID: mdl-36960857

ABSTRACT

Glioblastoma multiforme (GBM; World Health Organization grade IV) is one of the most common and aggressive malignant brain tumors and has no effective treatment. Therefore, elucidation of the molecular mechanism of glioma development is very important for finding new therapeutic strategies. The present study evaluated the expression level of Vav guanine nucleotide exchange factor 3 (VAV3) using bioinformatics analysis and demonstrated that VAV3 was overexpressed in human glioblastoma and associated with patient survival. Knock down of VAV3 using shRNA in glioblastoma cells significantly inhibited glioblastoma cell migration, invasion and proliferation. Furthermore, downregulation of VAV3 expression inhibited the stem cell self­renewal capacity and decreased the expression levels of the stem cell markers Nestin and Sox2. Bioinformatic analysis demonstrated that VAV3 was a target gene of miR­218. Furthermore, overexpression of VAV3 markedly reversed the tumor suppressor effect of miR­218 in glioblastoma cell. These findings suggested that VAV3 could be a potential biomarker and therapeutic target for glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , MicroRNAs , Humans , Glioblastoma/pathology , Cell Self Renewal/genetics , Cell Line, Tumor , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , Cell Proliferation/genetics , Brain Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/therapeutic use , Gene Expression Regulation, Neoplastic , Cell Movement/genetics
10.
Gene ; 863: 147306, 2023 May 05.
Article in English | MEDLINE | ID: mdl-36813057

ABSTRACT

The role of the Arhgef1 as a RhoA-specific guanine nucleotide exchange factor has been widely investigated in the immune system. Our previous findings reveal that Arhgef 1 is highly expressed in neural stem cells (NSCs) and controls the process of neurite formation. However, the functional role of Arhgef 1 in NSCs remains poorly understood. In order to investigate the role of Arhgef 1 in NSCs, Arhgef 1 expression in NSCs was reduced by using lentivirus-mediated short hairpin RNA interference. Our results indicate that down-regulated expression of Arhgef 1 reduced the self-renewal, proliferation capacity of NSCs and affect cell fate determination. In addition, the comparative transcriptome analysis from RNA-seq data determines the mechanisms of deficits in Arhgef 1 knockdown NSCs. Altogether, our present studies show that Arhgef 1 down-regulation leads to interruption of the cell cycle procession. The importance of Arhgef 1 for regulating self-renewal, proliferation and differentiation in NSCs is reported for the first time.


Subject(s)
Cell Self Renewal , Neural Stem Cells , Cell Self Renewal/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Cell Differentiation , Neural Stem Cells/metabolism , Neurogenesis
11.
Cell Death Differ ; 30(1): 184-194, 2023 01.
Article in English | MEDLINE | ID: mdl-36114296

ABSTRACT

Alternative splicing (AS) is tightly regulated during cell differentiation and development. AS events are prevalent in the testis, but the splicing regulation in spermatogenesis remains unclear. Here we report that the spliceosome component Bud31 plays a crucial role during spermatogenesis in mice. Germ cell-specific knockout of Bud31 led to loss of spermatogonia and to male infertility. We further demonstrate that Bud31 is required for both spermatogonial stem cell pool maintenance and the initiation of spermatogenesis. SMART-seq revealed that deletion of Bud31 in germ cells causes widespread exon-skipping and intron retention. Particularly, we identified Cdk2 as one of the direct splicing targets of Bud31, knockout of Bud31 resulted in retention of the first intron of Cdk2, which led to a decrease in Cdk2 expression. Our findings suggest that Bud31-mediated AS within spermatogonial stem cells regulates the self-renewal and differentiation of male germ cells in mammals.


Subject(s)
Cell Self Renewal , Spermatogonia , Male , Animals , Mice , Spermatogonia/physiology , Cell Self Renewal/genetics , Alternative Splicing/genetics , Testis/metabolism , Spermatogenesis/genetics , Cell Differentiation/genetics , Mammals
12.
Biochem Biophys Res Commun ; 641: 1-9, 2023 01 22.
Article in English | MEDLINE | ID: mdl-36516479

ABSTRACT

Stem cell niche provides extrinsic signals to maintain stem cell renewal or initiate cell differentiation. Drosophila niche is composed of somatic terminal filament cells, cap cells and escort cells. However, the underlying mechanism for the development of stem cell niche remains largely unclear. Here we found that the expression of a zinc transporter Catsup is essential for ovary morphogenesis. Catsup knockdown in escort cells results in defects of niche establishment and germline stem cells self-renewal. These defects could be modified by altered expression of genes involved in zinc metabolism or intervention of dietary zinc levels. Further studies indicated that Catsup RNAi affected adult ovary morphogenesis by suppressing Notch signaling. Lastly, we demonstrated that the defects of Catsup RNAi could be restored by overexpression of heat shock cognate protein 70 (Hsc70). These findings expand our understanding of the mechanisms controlling adult oogenesis and niche establishment in Drosophila.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Female , Drosophila/metabolism , Ovary/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Cell Self Renewal/genetics , Stem Cell Niche/genetics , Cell Differentiation , Stem Cells/metabolism , Germ Cells , Drosophila melanogaster/metabolism
13.
Gene Expr Patterns ; 47: 119302, 2023 03.
Article in English | MEDLINE | ID: mdl-36516960

ABSTRACT

Transcriptional enhanced associate domain (TEAD) transcription factors play important roles in embryonic stem cell (ESC) renewal and differentiation. Four TEAD transcription factors (Tead1, Tead2, Tead3 and Tead4) and their various splice variants have been discovered in mice, but the expression pattern of them during pluripotency state transition is unclear. Here, we investigated the expression of TEADs and their splice variants in mouse ESCs at different pluripotent/differentiating states and adult mouse tissues. Our results preliminarily revealed the diversity and heterogeneity of TEAD family, which is helpful for understanding their overlapping and distinctive functions. Furthermore, a novel splice variant of Tead1 was identified and named Tead1 isoform 4.


Subject(s)
Cell Differentiation , Cell Self Renewal , Mouse Embryonic Stem Cells , TEA Domain Transcription Factors , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Gene Expression Profiling , Mouse Embryonic Stem Cells/metabolism , RNA Isoforms/genetics , RNA Splicing/genetics , TEA Domain Transcription Factors/genetics , TEA Domain Transcription Factors/metabolism , Alternative Splicing/genetics , Cells, Cultured
14.
Nat Commun ; 13(1): 6107, 2022 10 16.
Article in English | MEDLINE | ID: mdl-36245043

ABSTRACT

Acute myeloid leukemia (AML) is maintained by self-renewing leukemic stem cells (LSCs). A fundamental problem in treating AML is that conventional therapy fails to eliminate LSCs, which can reinitiate leukemia. Heat shock transcription factor 1 (HSF1), a central regulator of the stress response, has emerged as an important target in cancer therapy. Using genetic Hsf1 deletion and a direct HSF1 small molecule inhibitor, we show that HSF1 is specifically required for the maintenance of AML, while sparing steady-state and stressed hematopoiesis. Mechanistically, deletion of Hsf1 dysregulates multifaceted genes involved in LSC stemness and suppresses mitochondrial oxidative phosphorylation through downregulation of succinate dehydrogenase C (SDHC), a direct HSF1 target. Forced expression of SDHC largely restores the Hsf1 ablation-induced AML developmental defect. Importantly, the growth and engraftment of human AML cells are suppressed by HSF1 inhibition. Our data provide a rationale for developing efficacious small molecules to specifically target HSF1 in AML.


Subject(s)
Cell Self Renewal , Leukemia, Myeloid, Acute , Humans , Cell Self Renewal/genetics , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Succinate Dehydrogenase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Leukemia ; 36(6): 1575-1584, 2022 06.
Article in English | MEDLINE | ID: mdl-35461365

ABSTRACT

Acute myeloid leukemia (AML) is characterized by poor clinical outcomes due to high rates of relapse following standard-of-care induction chemotherapy. While many pathogenic drivers have been described in AML, our understanding of the molecular mechanisms mediating chemotherapy resistance remains poor. Therefore, we sought to identify resistance genes to induction therapy in AML and elucidated ALOX5 as a novel mediator of resistance to anthracycline-based therapy. ALOX5 is transcriptionally upregulated in AML patient blasts in comparison to normal hematopoietic stem/progenitor cells (HSPCs) and ALOX5 mRNA, and protein expression is increased in response to induction therapy. In vitro, and in vivo genetic, and pharmacologic perturbation studies confirm that ALOX5 positively regulates the leukemogenic potential of AML LSCs, and its loss does not significantly affect the function of normal HSPCs. ALOX5 mediates resistance to daunorubicin (DNR) and promotes AML cell survival and maintenance through its leukotriene (LT) synthetic capacity, specifically via modulating the synthesis of LTB4 and its binding to LTB receptor (BLTR). Our study reveals a previously unrecognized role of LTs in AML pathogenesis and chemoresistance, whereby inhibition of ALOX5 mediated LTB4 synthesis and function could be combined with standard chemotherapy, to enhance the overall therapeutic efficacy in AML.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Antineoplastic Agents/therapeutic use , Cell Self Renewal/genetics , Daunorubicin/pharmacology , Daunorubicin/therapeutic use , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukotriene B4/metabolism , Leukotriene B4/therapeutic use , Neoplastic Stem Cells/pathology
16.
Cell Rep ; 38(10): 110481, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263585

ABSTRACT

Gene expression profiling and proteome analysis of normal and malignant hematopoietic stem cells (HSCs) point to shared core stemness properties. However, discordance between mRNA and protein signatures highlights an important role for post-transcriptional regulation by microRNAs (miRNAs) in governing this critical nexus. Here, we identify miR-130a as a regulator of HSC self-renewal and differentiation. Enforced expression of miR-130a impairs B lymphoid differentiation and expands long-term HSCs. Integration of protein mass spectrometry and chimeric AGO2 crosslinking and immunoprecipitation (CLIP) identifies TBL1XR1 as a primary miR-130a target, whose loss of function phenocopies miR-130a overexpression. Moreover, we report that miR-130a is highly expressed in t(8;21) acute myeloid leukemia (AML), where it is critical for maintaining the oncogenic molecular program mediated by the AML1-ETO complex. Our study establishes that identification of the comprehensive miRNA targetome within primary cells enables discovery of genes and molecular networks underpinning stemness properties of normal and leukemic cells.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , Cell Line, Tumor , Cell Self Renewal/genetics , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , MicroRNAs/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
17.
Blood Adv ; 6(4): 1095-1099, 2022 02 22.
Article in English | MEDLINE | ID: mdl-34516632

ABSTRACT

Mammalian GATA2 gene encodes a dual zinc finger transcription factor, which is essential for hematopoietic stem cell (HSC) generation in the aorta, gonad, mesonephros (AGM) region, HSC self-renewal, and specification of progenitor cell fates. Previously, we demonstrated that Gata2 expression in AGM is controlled by its intronic +9.5 enhancer. Gata2 +9.5 deficiency removes the E-box motif and the GATA site and depletes fetal liver HSCs. However, whether this enhancer has an essential role in regulating adult hematopoiesis has not been established. Here, we evaluate Gata2 +9.5 enhancer function in adult hematopoiesis. +9.5+/- bone marrow cells displayed reduced T cell reconstitution in a competitive transplant assay. Donor-derived analysis demonstrated a previously unrecognized function of the +9.5 enhancer in T cell development at the lymphoid-primed multipotent progenitor stage. Moreover, +9.5+/- adult HSCs displayed increased apoptosis and reduced long-term self-renewal capability in comparison with wild-type (WT) HSCs. These phenotypes were more moderate than those of Gata2+/- HSCs. Consistent with the phenotypic characterization, Gata2 expression in +9.5+/- LSKs was moderately higher than that in Gata2+/- LSKs, but lower than that in WT LSKs. Our data suggest that +9.5 deficiency compromises, without completely abrogating, Gata2 expression in adult HSCs.


Subject(s)
Hematopoiesis , Mesonephros , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mammals
18.
PLoS Genet ; 17(12): e1009609, 2021 12.
Article in English | MEDLINE | ID: mdl-34898607

ABSTRACT

How and when potential becomes restricted in differentiating stem cell daughters is poorly understood. While it is thought that signals from the niche are actively required to prevent differentiation, another model proposes that stem cells can reversibly transit between multiple states, some of which are primed, but not committed, to differentiate. In the Drosophila testis, somatic cyst stem cells (CySCs) generate cyst cells, which encapsulate the germline to support its development. We find that CySCs are maintained independently of niche self-renewal signals if activity of the PI3K/Tor pathway is inhibited. Conversely, PI3K/Tor is not sufficient alone to drive differentiation, suggesting that it acts to license cells for differentiation. Indeed, we find that the germline is required for differentiation of CySCs in response to PI3K/Tor elevation, indicating that final commitment to differentiation involves several steps and intercellular communication. We propose that CySC daughter cells are plastic, that their fate depends on the availability of neighbouring germ cells, and that PI3K/Tor acts to induce a primed state for CySC daughters to enable coordinated differentiation with the germline.


Subject(s)
Adult Stem Cells/cytology , Drosophila Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , TOR Serine-Threonine Kinases/genetics , Testis/growth & development , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Germ Cells/growth & development , Male , Signal Transduction/genetics , Stem Cell Niche/genetics , Testis/metabolism
19.
Cells ; 10(12)2021 11 23.
Article in English | MEDLINE | ID: mdl-34943786

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Mesenchymal Stem Cells/cytology , Regenerative Medicine , Amniotic Fluid/cytology , Cell Self Renewal/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Female , Humans , Mesenchymal Stem Cell Transplantation , Placenta/cytology , Placenta/transplantation , Pregnancy , Umbilical Cord/cytology , Umbilical Cord/transplantation
20.
Pancreas ; 50(9): 1260-1266, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34860809

ABSTRACT

OBJECTIVES: This study aimed to enhance the sensitivity of pancreatic ductal adenocarcinoma cells by microRNA-34a (miR-34a)-mediated targeting of Notch 1. METHODS: Cell viability was determined by using an MTT (3-(4,5)-dimethylthiahiazo(-2)-3,5-diphenytetrazoliumromide) assay. The expression levels of miR-34a and relevant mRNAs were determined using quantitative polymerase chain reaction. Protein levels were measured by Western blotting. Cellular stemness was assessed by cell invasiveness and sphere formation assays. A transplanted tumor model was established for in vivo experiments. RESULTS: MicroRNA-34a enhanced gemcitabine sensitivity both in vivo and in vitro. MicroRNA-34a suppressed the stemness and proliferation of pancreatic cancer stem cells. MicroRNA-34a directly associated with Notch 1, which lies upstream of epithelial-mesenchymal transition signaling pathways. CONCLUSIONS: MicroRNA-34a sensitized pancreatic cancer cells to gemcitabine treatment by inhibiting Notch 1 signaling in pancreatic cancer stem cells, indicating that miR-34a has the potential to be developed as a novel therapeutic agent for the treatment of gemcitabine-resistant pancreatic ductal adenocarcinoma cells.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Self Renewal/genetics , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Self Renewal/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Deoxycytidine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays/methods , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...