Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Exp Hematol ; 109: 35-44, 2022 05.
Article in English | MEDLINE | ID: mdl-35192914

ABSTRACT

The Polycomb complex protein Bmi1 is regarded as a master regulator of hematopoietic stem cells (HSCs). In the blood system, HSCs express Bmi1 most abundantly, and Bmi1 expression wanes as cells differentiate. Furthermore, Bmi1 has been found to be overexpressed in several hematologic cancers. Most studies exploring the normal role of Bmi1 in HSC biology have used loss-of-function models, which have established Bmi1 as an important regulator for HSC maintenance. Additionally, gain-of-function studies using retroviral and lentiviral approaches have observed increased self-renewal of Bmi1-transduced HSCs. However, the clinical and biological relevance of such studies is typically hampered by uncontrolled transgenic integration and supraphysiological expression levels. Here, we describe how we developed a novel tetracycline-inducible gain-of-function Bmi1 (iBmi1) transgenic mouse model. We found that Bmi1 induction had minor, if any, effects on steady-state hematopoiesis or after 5-fluorouracil-induced cytostatic stress. On the contrary, secondary transplantation of iBmi1 HSCs into wild-type recipients resulted in marked increases in the number and chimerism of HSCs. These data, in concert with previous loss-of-function studies, suggest that although endogenous Bmi1 levels are required and sufficient for normal HSC maintenance, the stabilization of these levels over time protects HSCs from transplantation-associated stress.


Subject(s)
Hematopoietic Stem Cell Transplantation , Proto-Oncogene Proteins , Animals , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mice , Mice, Transgenic , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
2.
Cell Rep ; 21(12): 3514-3523, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262330

ABSTRACT

The transcription factor hepatic leukemia factor (HLF) is strongly expressed in hematopoietic stem cells (HSCs) and is thought to influence both HSC self-renewal and leukemogenesis. However, the physiological role of HLF in hematopoiesis and HSC function is unclear. Here, we report that mice lacking Hlf are viable with essentially normal hematopoietic parameters, including an intact HSC pool during steady-state hematopoiesis. In contrast, when challenged through transplantation, Hlf-deficient HSCs showed an impaired ability to reconstitute hematopoiesis and became gradually exhausted upon serial transplantation. Transcriptional profiling of Hlf-deficient HSCs revealed changes associated with enhanced cellular activation, and cell-cycle analysis demonstrated a significant reduction of quiescent HSCs. Accordingly, toxic insults targeting dividing cells completely eradicated the HSC pool in Hlf-deficient mice. In summary, our findings point to HLF as a critical regulator of HSC quiescence and as an essential factor for maintaining the HSC pool during regeneration.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Animals , Apoptosis , Basic-Leucine Zipper Transcription Factors/genetics , Cells, Cultured , DNA Damage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Transcriptome
3.
Cell Rep ; 21(8): 2251-2263, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166614

ABSTRACT

A gradual restriction in lineage potential of multipotent stem/progenitor cells is a hallmark of adult hematopoiesis, but the underlying molecular events governing these processes remain incompletely understood. Here, we identified robust expression of the leukemia-associated transcription factor hepatic leukemia factor (Hlf) in normal multipotent hematopoietic progenitors, which was rapidly downregulated upon differentiation. Interference with its normal downregulation revealed Hlf as a strong negative regulator of lymphoid development, while remaining compatible with myeloid fates. Reciprocally, we observed rapid lymphoid commitment upon reduced Hlf activity. The arising phenotypes resulted from Hlf binding to active enhancers of myeloid-competent cells, transcriptional induction of myeloid, and ablation of lymphoid gene programs, with Hlf induction of nuclear factor I C (Nfic) as a functionally relevant target gene. Thereby, our studies establish Hlf as a key regulator of the earliest lineage-commitment events at the transition from multipotency to lineage-restricted progeny, with implications for both normal and malignant hematopoiesis.


Subject(s)
Cell Lineage/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Leukemia/metabolism , Multipotent Stem Cells/cytology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation/physiology , Lymphopoiesis/physiology , Mice , Myeloid Cells/metabolism
4.
Exp Hematol ; 56: 1-6, 2017 12.
Article in English | MEDLINE | ID: mdl-28943295

ABSTRACT

The late stages of life, in most species including humans, are associated with a decline in the overall maintenance and health of the organism. This applies also to the hematopoietic system, where aging is not only associated with an increased predisposition for hematological malignancies, but also identified as a strong comorbidity factor for other diseases. Research during the last two decades has proposed that alterations at the level of hematopoietic stem cells (HSCs) might be a root cause for the hematological changes observed with age. However, the recent realization that not all HSCs are alike with regard to fundamental stem cell properties such as self-renewal and lineage potential has several implications for HSC aging, including the synchrony and the stability of the aging HSC state. To approach HSC aging from a clonal perspective, we recently took advantage of technical developments in cellular barcoding and combined this with the derivation of induced pluripotent stem cells (iPSCs). This allowed us to selectively approach HSCs functionally affected by age. The finding that such iPSCs were capable of fully regenerating multilineage hematopoiesis upon morula/blastocyst complementation provides compelling evidence that many aspects of HSC aging can be reversed, which indicates that a central mechanism underlying HSC aging is a failure to uphold the epigenomes associated with younger age. Here we discuss these findings in the context of the underlying causes that might influence HSC aging and the requirements and prospects for restoration of the aging HSC epigenome.


Subject(s)
Cellular Senescence , Epigenesis, Genetic , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Animals , Humans , Induced Pluripotent Stem Cells/metabolism
5.
Nat Commun ; 8: 14533, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28224997

ABSTRACT

Ageing associates with significant alterations in somatic/adult stem cells and therapies to counteract these might have profound benefits for health. In the blood, haematopoietic stem cell (HSC) ageing is linked to several functional shortcomings. However, besides the recent realization that individual HSCs might be preset differentially already from young age, HSCs might also age asynchronously. Evaluating the prospects for HSC rejuvenation therefore ultimately requires approaching those HSCs that are functionally affected by age. Here we combine genetic barcoding of aged murine HSCs with the generation of induced pluripotent stem (iPS) cells. This allows us to specifically focus on aged HSCs presenting with a pronounced lineage skewing, a hallmark of HSC ageing. Functional and molecular evaluations reveal haematopoiesis from these iPS clones to be indistinguishable from that associating with young mice. Our data thereby provide direct support to the notion that several key functional attributes of HSC ageing can be reversed.


Subject(s)
Aging/physiology , Cell Lineage , Cellular Senescence , Clone Cells/cytology , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Lineage/genetics , Cellular Reprogramming/genetics , Cellular Senescence/genetics , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
6.
Stem Cells Transl Med ; 4(2): 186-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25548388

ABSTRACT

Because of the continuous increases in lifetime expectancy, the incidence of age-related diseases will, unless counteracted, represent an increasing problem at both the individual and socioeconomic levels. Studies on the processes of blood cell formation have revealed several shortcomings as a consequence of chronological age. They include a reduced ability to mount adaptive immune responses and a blood cell composition skewed toward myeloid cells, with the latter coinciding with a dramatically increased incidence of myelogenous diseases, including cancer. Conversely, the dominant forms of acute leukemia affecting children associate with the lymphoid lineages. A growing body of evidence has suggested that aging of various organs and cellular systems, including the hematopoietic system, associates with a functional demise of tissue-resident stem cell populations. Mechanistically, DNA damage and/or altered transcriptional landscapes appear to be major drivers of the hematopoietic stem cell aging state, with recent data proposing that stem cell aging phenotypes are characterized by at least some degree of reversibility. These findings suggest the possibility of rejuvenating, or at least dampening, stem cell aging phenotypes in the elderly for therapeutic benefit.


Subject(s)
Hematopoietic Stem Cells/metabolism , Rejuvenation , Animals , Hematologic Diseases/therapy , Hematopoietic Stem Cell Transplantation , Humans , Neoplasms/therapy
7.
Cell Rep ; 9(4): 1246-55, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456127

ABSTRACT

Studies of developmental pathways of hematopoietic stem cells (HSCs) have defined lineage relationships throughout the blood system. This is relevant to acute myeloid leukemia (AML), where aggressiveness and therapeutic responsiveness can be influenced by the initial stage of transformation. To address this, we generated a mouse model in which the mixed-lineage leukemia/eleven-nineteen-leukemia (MLL-ENL) transcription factor can be conditionally activated in any cell type. We show that AML can originate from multiple hematopoietic progenitor subsets with granulocytic and monocytic potential, and that the normal developmental position of leukemia-initiating cells influences leukemic development. However, disease failed to arise from HSCs. Although it maintained or upregulated the expression of target genes associated with leukemic development, MLL-ENL dysregulated the proliferative and repopulating capacity of HSCs. Therefore, the permissiveness for development of AML may be associated with a narrower window of differentiation than was previously appreciated, and hijacking the self-renewal capacity of HSCs by a potent oncogene is insufficient for leukemic development.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cytoprotection , Hematopoietic Stem Cells/cytology , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Differentiation/drug effects , Cytoprotection/drug effects , Disease Models, Animal , Doxycycline/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Progenitor Cells/pathology , Reproducibility of Results , Transcription, Genetic/drug effects
8.
Cell Stem Cell ; 14(5): 555-6, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24792111

ABSTRACT

Obtaining sufficient numbers of immunologically matched hematopoietic stem cells (HSCs) poses a major clinical hurdle in bone marrow transplantation therapies. In a recent study in Cell, Riddell et al. (2014) generate induced HSCs from differentiated blood cells, which may serve as a potential solution to this clinical challenge.


Subject(s)
Cellular Reprogramming , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism , Animals
9.
Stem Cells ; 32(5): 1173-82, 2014 May.
Article in English | MEDLINE | ID: mdl-24446123

ABSTRACT

It has become increasingly clear that several age-associated pathologies associate with mutations in the mitochondrial genome. Experimental modeling of such events has revealed that acquisition of mitochondrial DNA (mtDNA) damage can impair respiratory function and, as a consequence, can lead to widespread decline in cellular function. This includes premature aging syndromes. By taking advantage of a mutator mouse model with an error-prone mtDNA polymerase, we here investigated the impact of an established mtDNA mutational load with regards to the generation, maintenance, and differentiation of induced pluripotent stem (iPS) cells. We demonstrate that somatic cells with a heavy mtDNA mutation burden were amenable for reprogramming into iPS cells. However, mutator iPS cells displayed delayed proliferation kinetics and harbored extensive differentiation defects. While mutator iPS cells had normal ATP levels and glycolytic activity, the induction of differentiation coincided with drastic decreases in ATP production and a hyperactive glycolysis. These data demonstrate the differential requirements of mitochondrial integrity for pluripotent stem cell self-renewal versus differentiation and highlight the relevance of assessing the mitochondrial genome when aiming to generate iPS cells with robust differentiation potential.


Subject(s)
Cell Differentiation/genetics , DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , Mutation , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Cellular Reprogramming/genetics , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , Glycolysis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Electron, Transmission , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Octamer Transcription Factor-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics
10.
Blood ; 121(21): 4257-64, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23476050

ABSTRACT

Aging of hematopoietic stem cells (HSCs) leads to several functional changes, including alterations affecting self-renewal and differentiation. Although it is well established that many of the age-induced changes are intrinsic to HSCs, less is known regarding the stability of this state. Here, we entertained the hypothesis that HSC aging is driven by the acquisition of permanent genetic mutations. To examine this issue at a functional level in vivo, we applied induced pluripotent stem (iPS) cell reprogramming of aged hematopoietic progenitors and allowed the resulting aged-derived iPS cells to reform hematopoiesis via blastocyst complementation. Next, we functionally characterized iPS-derived HSCs in primary chimeras and after the transplantation of re-differentiated HSCs into new hosts, the gold standard to assess HSC function. Our data demonstrate remarkably similar functional properties of iPS-derived and endogenous blastocyst-derived HSCs, despite the extensive chronological and proliferative age of the former. Our results, therefore, favor a model in which an underlying, but reversible, epigenetic component is a hallmark of HSC aging.


Subject(s)
Cell Differentiation/physiology , Cellular Senescence/physiology , Epigenesis, Genetic/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cellular Senescence/genetics , Gene Expression Regulation, Developmental/physiology , Genome-Wide Association Study , Mice , Mice, Inbred C57BL , Telomere/genetics , Totipotent Stem Cells/cytology , Totipotent Stem Cells/physiology , Transcription, Genetic/physiology
11.
Aging Cell ; 11(6): 1128-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22809070

ABSTRACT

Aging causes profound effects on the hematopoietic stem cell (HSC) pool, including an altered output of mature progeny and enhanced self-propagation of repopulating-defective HSCs. An important outstanding question is whether HSCs can be protected from aging. The signal adaptor protein LNK negatively regulates hematopoiesis at several cellular stages. It has remained unclear how the enhanced sensitivity to cytokine signaling caused by LNK deficiency affects hematopoiesis upon aging. Our findings demonstrate that aged LNK-/- HSCs displayed a robust overall reconstitution potential and gave rise to a hematopoietic system with a balanced lineage distribution. Although aged LNK-/- HSCs displayed a distinct molecular profile in which reduced proliferation was central, little or no difference in the proliferation of aged LNK-/- HSCs was observed after transplantation when compared to aged WT HSCs. This coincided with equal telomere maintenance in WT and LNK-/- HSCs. Collectively, our studies suggest that enhanced cytokine signaling can counteract functional age-related HSC decline.


Subject(s)
Aging/genetics , Cytokines/genetics , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Adaptor Proteins, Signal Transducing , Aging/metabolism , Animals , Cell Proliferation , Cytokines/biosynthesis , Gene Expression Regulation , Gene Knockdown Techniques , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Homeostasis , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins , Mice , Signal Transduction , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis
12.
Cell Stem Cell ; 8(5): 499-510, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21549326

ABSTRACT

Somatic stem cells mediate tissue maintenance for the lifetime of an organism. Despite the well-established longevity that is a prerequisite for such function, accumulating data argue for compromised stem cell function with age. Identifying the mechanisms underlying age-dependent stem cell dysfunction is therefore key to understanding the aging process. Here, using a model carrying a proofreading-defective mitochondrial DNA polymerase, we demonstrate hematopoietic defects reminiscent of premature HSC aging, including anemia, lymphopenia, and myeloid lineage skewing. However, in contrast to physiological stem cell aging, rapidly accumulating mitochondrial DNA mutations had little functional effect on the hematopoietic stem cell pool, and instead caused distinct differentiation blocks and/or disappearance of downstream progenitors. These results show that intact mitochondrial function is required for appropriate multilineage stem cell differentiation, but argue against mitochondrial DNA mutations per se being a primary driver of somatic stem cell aging.


Subject(s)
Aging, Premature/genetics , Cellular Senescence/physiology , DNA-Directed DNA Polymerase/metabolism , Hematopoietic Stem Cells/metabolism , Mitochondria/metabolism , Adult Stem Cells/physiology , Aging, Premature/physiopathology , Anemia , Animals , Cells, Cultured , Chimera , DNA Polymerase gamma , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Gene Expression Profiling , Hematopoiesis/genetics , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Lymphopenia , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL
...