Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 24(5): e55373, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36943011

ABSTRACT

Upon ex vivo culture, hematopoietic stem cells (HSCs) quickly lose potential and differentiate into progenitors. The identification of culture conditions that maintain the potential of HSCs ex vivo is therefore of high clinical interest. Here, we demonstrate that the potential of murine and human HSCs is maintained when cultivated for 2 days ex vivo at a pH of 6.9, in contrast to cultivation at the commonly used pH of 7.4. When cultivated at a pH of 6.9, HSCs remain smaller, less metabolically active, less proliferative and show enhanced reconstitution ability upon transplantation compared to HSC cultivated at pH 7.4. HSCs kept at pH 6.9 show an attenuated polyamine pathway. Pharmacological inhibition of the polyamine pathway in HSCs cultivated at pH 7.4 with DFMO mimics phenotypes and potential of HSCs cultivated at pH 6.9. Ex vivo exposure to a pH of 6.9 is therefore a positive regulator of HSC function by reducing polyamines. These findings might improve HSC short-term cultivation protocols for transplantation and gene therapy interventions.


Subject(s)
Hematopoietic Stem Cells , Humans , Mice , Animals , Hematopoietic Stem Cells/metabolism , Hydrogen-Ion Concentration
2.
Life Sci Alliance ; 5(8)2022 08.
Article in English | MEDLINE | ID: mdl-35487692

ABSTRACT

Upon aging, the function of the intestinal epithelium declines with a concomitant increase in aging-related diseases. ISCs play an important role in this process. It is known that ISC clonal dynamics follow a neutral drift model. However, it is not clear whether the drift model is still valid in aged ISCs. Tracking of clonal dynamics by clonal tracing revealed that aged crypts drift into monoclonality substantially faster than young ones. However, ISC tracing experiments, in vivo and ex vivo, implied a similar clonal expansion ability of both young and aged ISCs. Single-cell RNA sequencing for 1,920 high Lgr5 ISCs from young and aged mice revealed increased heterogeneity among subgroups of aged ISCs. Genes associated with cell adhesion were down-regulated in aged ISCs. ISCs of aged mice indeed show weaker adhesion to the matrix. Simulations applying a single cell-based model of the small intestinal crypt demonstrated an accelerated clonal drift at reduced adhesion strength, implying a central role for reduced adhesion for affecting clonal dynamics upon aging.


Subject(s)
Intestines , Stem Cells , Animals , Cells, Cultured , Ileum , Intestinal Mucosa/metabolism , Mice , Stem Cells/metabolism
3.
Leukemia ; 35(6): 1751-1762, 2021 06.
Article in English | MEDLINE | ID: mdl-33077869

ABSTRACT

Shwachman-Diamond syndrome (SDS) is a bone marrow failure (BMF) syndrome associated with an increased risk of myelodysplasia and leukemia. The molecular mechanisms of SDS are not fully understood. We report that primitive hematopoietic cells from SDS patients present with a reduced activity of the small RhoGTPase Cdc42 and concomitantly a reduced frequency of HSCs polar for polarity proteins. The level of apolarity of SDS HSCs correlated with the magnitude of HSC depletion in SDS patients. Importantly, exogenously provided Wnt5a or GDF11 that elevates the activity of Cdc42 restored polarity in SDS HSCs and increased the number of HSCs in SDS patient samples in surrogate ex vivo assays. Single cell level RNA-Seq analyses of SDS HSCs and daughter cells demonstrated that SDS HSC treated with GDF11 are transcriptionally more similar to control than to SDS HSCs. Treatment with GDF11 reverted pathways in SDS HSCs associated with rRNA processing and ribosome function, but also viral infection and immune function, p53-dependent DNA damage, spindle checkpoints, and metabolism, further implying a role of these pathways in HSC failure in SDS. Our data suggest that HSC failure in SDS is driven at least in part by low Cdc42 activity in SDS HSCs. Our data thus identify novel rationale approaches to attenuate HSCs failure in SDS.


Subject(s)
Bone Marrow Cells/cytology , Cell Polarity , Hematopoietic Stem Cells/cytology , Shwachman-Diamond Syndrome/prevention & control , cdc42 GTP-Binding Protein/metabolism , Bone Marrow Cells/metabolism , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Growth Differentiation Factors/chemistry , Growth Differentiation Factors/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Prognosis , Shwachman-Diamond Syndrome/etiology , Shwachman-Diamond Syndrome/metabolism , Shwachman-Diamond Syndrome/pathology , Wnt-5a Protein/chemistry , Wnt-5a Protein/metabolism , cdc42 GTP-Binding Protein/chemistry
4.
Aging Cell ; 19(9): e13208, 2020 09.
Article in English | MEDLINE | ID: mdl-32755011

ABSTRACT

Cdc42 is a small RhoGTPase regulating multiple functions in eukaryotic cells. The activity of Cdc42 is significantly elevated in several tissues of aged mice, while the Cdc42 gain-of-activity mouse model presents with a premature aging-like phenotype and with decreased lifespan. These data suggest a causal connection between elevated activity of Cdc42, aging, and reduced lifespan. Here, we demonstrate that systemic treatment of aged (75-week-old) female C57BL/6 mice with a Cdc42 activity-specific inhibitor (CASIN) for 4 consecutive days significantly extends average and maximum lifespan. Moreover, aged CASIN-treated animals displayed a youthful level of the aging-associated cytokines IL-1ß, IL-1α, and INFγ in serum and a significantly younger epigenetic clock as based on DNA methylation levels in blood cells. Overall, our data show that systemic administration of CASIN to reduce Cdc42 activity in aged mice extends murine lifespan.


Subject(s)
Cytokines/metabolism , cdc42 GTP-Binding Protein/genetics , Aging , Animals , Drosophila Proteins , Female , Integrin alpha Chains , Longevity , Mice , Mice, Inbred C57BL
6.
PLoS Biol ; 16(9): e2003389, 2018 09.
Article in English | MEDLINE | ID: mdl-30235201

ABSTRACT

Hematopoietic stem cells (HSCs) balance self-renewal and differentiation to maintain homeostasis. With aging, the frequency of polar HSCs decreases. Cell polarity in HSCs is controlled by the activity of the small RhoGTPase cell division control protein 42 (Cdc42). Here we demonstrate-using a comprehensive set of paired daughter cell analyses that include single-cell 3D confocal imaging, single-cell transplants, single-cell RNA-seq, and single-cell transposase-accessible chromatin sequencing (ATAC-seq)-that the outcome of HSC divisions is strongly linked to the polarity status before mitosis, which is in turn determined by the level of the activity Cdc42 in stem cells. Aged apolar HSCs undergo preferentially self-renewing symmetric divisions, resulting in daughter stem cells with reduced regenerative capacity and lymphoid potential, while young polar HSCs undergo preferentially asymmetric divisions. Mathematical modeling in combination with experimental data implies a mechanistic role of the asymmetric sorting of Cdc42 in determining the potential of daughter cells via epigenetic mechanisms. Therefore, molecules that control HSC polarity might serve as modulators of the mode of stem cell division regulating the potential of daughter cells.


Subject(s)
Cell Division/genetics , Cellular Senescence/genetics , Epigenesis, Genetic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Aging/metabolism , Animals , Asymmetric Cell Division/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Aggregation , Cell Lineage/drug effects , Cell Polarity/drug effects , Chromatin , Mice, Inbred C57BL , Transcriptome/genetics , Wnt-5a Protein/pharmacology , cdc42 GTP-Binding Protein/metabolism
7.
Cell Rep ; 18(11): 2608-2621, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297666

ABSTRACT

Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.


Subject(s)
Cellular Senescence , Intestine, Small/cytology , Stem Cells/cytology , Stem Cells/metabolism , Wnt Signaling Pathway , Animals , Biomarkers/metabolism , Cell Count , Cell Proliferation , Female , Mice , Organoids/cytology , Regeneration , Stem Cell Niche
8.
Nature ; 503(7476): 392-6, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24141946

ABSTRACT

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Subject(s)
Cellular Senescence , Hematopoietic Stem Cells/cytology , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Polarity , Female , Haploinsufficiency , Male , Mice , Mice, Inbred C57BL , Phenotype , Rejuvenation , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein , cdc42 GTP-Binding Protein/metabolism
9.
Nat Med ; 18(7): 1123-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22729286

ABSTRACT

Tissue damage induced by ionizing radiation in the hematopoietic and gastrointestinal systems is the major cause of lethality in radiological emergency scenarios and underlies some deleterious side effects in patients undergoing radiation therapy. The identification of target-specific interventions that confer radiomitigating activity is an unmet challenge. Here we identify the thrombomodulin (Thbd)-activated protein C (aPC) pathway as a new mechanism for the mitigation of total body irradiation (TBI)-induced mortality. Although the effects of the endogenous Thbd-aPC pathway were largely confined to the local microenvironment of Thbd-expressing cells, systemic administration of soluble Thbd or aPC could reproduce and augment the radioprotective effect of the endogenous Thbd-aPC pathway. Therapeutic administration of recombinant, soluble Thbd or aPC to lethally irradiated wild-type mice resulted in an accelerated recovery of hematopoietic progenitor activity in bone marrow and a mitigation of lethal TBI. Starting infusion of aPC as late as 24 h after exposure to radiation was sufficient to mitigate radiation-induced mortality in these mice. These findings suggest that pharmacologic augmentation of the activity of the Thbd-aPC pathway by recombinant Thbd or aPC might offer a rational approach to the mitigation of tissue injury and lethality caused by ionizing radiation.


Subject(s)
Protein C/antagonists & inhibitors , Radiation Injuries/prevention & control , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Thrombomodulin/antagonists & inhibitors , Animals , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Protein C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Injuries/genetics , Radiation Injuries/pathology , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Receptors, Thrombin , Survival Analysis , Thrombomodulin/genetics , Thrombomodulin/metabolism , Whole-Body Irradiation
10.
Nat Med ; 16(10): 1141-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20871610

ABSTRACT

Mobilization of hematopoietic stem and progenitor cells (HSPCs) from bone marrow into peripheral blood by the cytokine granulocyte colony-stimulating factor (G-CSF) has become the preferred source of HSPCs for stem cell transplants. However, G-CSF fails to mobilize sufficient numbers of stem cells in up to 10% of donors, precluding autologous transplantation in those donors or substantially delaying transplant recovery time. Consequently, new regimens are needed to increase the number of stem cells in peripheral blood upon mobilization. Using a forward genetic approach in mice, we mapped the gene encoding the epidermal growth factor receptor (Egfr) to a genetic region modifying G-CSF-mediated HSPC mobilization. Amounts of EGFR in HSPCs inversely correlated with the cells' ability to be mobilized by G-CSF, implying a negative role for EGFR signaling in mobilization. In combination with G-CSF treatment, genetic reduction of EGFR activity in HSPCs (in waved-2 mutant mice) or treatment with the EGFR inhibitor erlotinib increased mobilization. Increased mobilization due to suppression of EGFR activity correlated with reduced activity of cell division control protein-42 (Cdc42), and genetic Cdc42 deficiency in vivo also enhanced G-CSF-induced mobilization. Our findings reveal a previously unknown signaling pathway regulating stem cell mobilization and provide a new pharmacological approach for improving HSPC mobilization and thereby transplantation outcomes.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Signal Transduction , Animals , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , cdc42 GTP-Binding Protein/physiology
11.
Methods Mol Biol ; 506: 423-36, 2009.
Article in English | MEDLINE | ID: mdl-19110642

ABSTRACT

Maintaining the stability of the genome is critical to cell survival and normal cell growth. Genetic modification of hematopoietic cells might bear an inherent increased risk for the accumulation of DNA mutations. It frequently requires cultivation of the cells under super-physiological oxygen levels, which can result in increased oxidative damage, as well as under super-physiological concentrations of cytokines, which might interfere with DNA-damage checkpoint activation and by this means might result in an increased mutational load. We describe here a protocol for monitoring the frequency of DNA mutations in bone marrow cells post transduction or upon selection either in vitro or in vivo based on the lacZ-plasmid (pUR288) transgenic mouse (small blue mouse) mutation indicator strain.


Subject(s)
Genomics , Hematopoietic Stem Cells/metabolism , Mutation , Animals , Base Sequence , DNA Primers , Electroporation , Gene Transfer Techniques , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymerase Chain Reaction
12.
J Clin Invest ; 117(11): 3283-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17960249

ABSTRACT

The molecular pathogenesis of the myeloid leukemias that frequently occur in patients with Fanconi anemia (FA) is not well defined. Hematopoietic stem cells bearing inactivating mutations of FA complementation group C (FANCC) are genetically unstable and hypersensitive to apoptotic cytokine cues including IFN-gamma and TNF-alpha, but neoplastic stem cell clones that arise frequently in vivo are resistant to these cytokines. Reasoning that the combination of genetic instability and cytokine hypersensitivity might create an environment supporting the emergence of leukemic stem cells, we tested the leukemia-promoting effects of TNF-alpha in murine stem cells. TNF-alpha exposure initially profoundly inhibited the growth of Fancc-/- stem cells. However, longer-term exposure of these cells promoted the outgrowth of cytogenetically abnormal clones that, upon transplantation into congenic WT mice, led to acute myelogenous leukemia. TNF-alpha induced ROS-dependent genetic instability in Fancc-/- but not in WT cells. The leukemic clones were TNF-alpha resistant but retained their characteristic hypersensitivity to mitomycin C and exhibited high levels of chromosomal instability. Expression of FANCC cDNA in Fancc-/- stem cells protected them from TNF-alpha-induced clonal evolution. We conclude that TNF-alpha exposure creates an environment in which somatically mutated preleukemic stem cell clones are selected and from which unaltered TNF-alpha-hypersensitive Fancc-/- stem cells are purged.


Subject(s)
Cell Proliferation , Fanconi Anemia Complementation Group C Protein/immunology , Fanconi Anemia/immunology , Hematopoietic Stem Cells/physiology , Tumor Necrosis Factor-alpha/immunology , Animals , Apoptosis , Cell Differentiation/physiology , Chromosome Aberrations , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Genetic Complementation Test , Hematopoietic Stem Cells/cytology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Stem Cell Transplantation , Survival Rate
13.
Blood ; 108(7): 2190-7, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16741255

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) are located in the bone marrow in close association with a highly organized 3-dimensional structure formed by stroma cells, referred to as the niche. Mobilization of HSPCs from bone marrow to peripheral blood in response to granulocyte colony-stimulating factor (G-CSF) requires de-adhesion of HSPCs from the niche. The influence of aging of HSPCs on cell-stroma interactions has not been determined in detail. Using a mouse model of G-CSF-induced mobilization, we demonstrated that the ability to mobilize hematopoietic stem cells is approximately 5-fold greater in aged mice. Competitive mobilization experiments confirmed that enhanced mobilization ability was intrinsic to the stem cell. Enhanced mobilization efficiency of primitive hematopoietic cells from aged mice correlated with reduced adhesion of hematopoietic progenitor cells to stroma and with elevated levels of GTP-bound Cdc42. These results might indicate that stroma-stem cell interactions are dynamic over a lifetime and result in physiologically relevant changes in the biology of primitive hematopoietic cells with age.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Aging , Animals , Cell Adhesion , Cell Movement , Granulocyte Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Stem Cells/cytology , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
15.
Am J Physiol Heart Circ Physiol ; 286(3): H1146-53, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14630633

ABSTRACT

We recently developed a mouse model with a single functional allele of Serca2 (Serca2+/-) that shows impaired cardiac contractility and relaxation without overt heart disease. The goal of this study was to test the hypothesis that chronic reduction in sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA)2 levels in combination with an increased hemodynamic load will result in an accelerated pathway to heart failure. Age-matched wild-type and Serca2+/- mice were subjected to 10 wk of pressure overload via transverse aortic coarctation surgery. Cardiac hypertrophy and heart failure were assessed by echocardiography, gravimetry/histology, hemodynamics, and Western blotting analyses. Our results showed that approximately 64% of coarcted Serca2+/- mice were in heart failure compared with 0% of coarcted wild-type mice (P < 0.05). Overall, morbidity and mortality were greatly increased in Serca2+/- mice under pressure overload. Echocardiography assessment revealed a significant increase in left ventricular (LV) mass, and LV hypertrophy in coarcted Serca2+/- mice converted from a concentric to an eccentric pattern, similar to that seen in human heart failure. Coarcted Serca2+/- mice had decreased contractile/systolic and relaxation/diastolic performance and/or function compared with coarcted wild-type mice (P < 0.05), despite a similar duration and degree of pressure overload. SERCA2a protein levels were significantly reduced (>50%) in coarcted Serca2+/- mice compared with noncoarcted and coarcted wild-type mice. Our findings suggest that reduction in SERCA2 levels in combination with an increased hemodynamic load results in an accelerated pathway to heart failure.


Subject(s)
Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Heart Failure/metabolism , Heart Failure/physiopathology , Animals , Calcium/metabolism , Cardiac Catheterization , Diastole , Echocardiography , Female , Heart Failure/diagnostic imaging , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Knockout , Phenotype , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Systole
16.
Am J Physiol Heart Circ Physiol ; 283(3): H958-65, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181124

ABSTRACT

In this study we evaluated the contractile characteristics of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA)1a-expressing hearts ex vivo and in vivo and in particular their response to beta-adrenergic stimulation. Analysis of isolated, work-performing hearts revealed that transgenic (TG) hearts develop much higher maximal rates of contraction and relaxation than wild-type (WT) hearts. Addition of isoproterenol only moderately increased the maximal rate of relaxation (+20%) but did not increase contractility or decrease relaxation time in TG hearts. Perfusion with varied buffer Ca(2+) concentrations indicated an altered dose response to Ca(2+). In vivo TG hearts displayed fairly higher maximal rates of contraction (+ 25%) but unchanged relaxation parameters and a blunted but significant response to dobutamine. Our study also shows that the phospholamban (PLB) level was decreased (-40%) and its phosphorylation status modified in TG hearts. This study clearly demonstrates that increases in SERCA protein level alter the beta-adrenergic response and affect the phosphorylation of PLB. Interestingly, the overall cardiac function in the live animal is only slightly enhanced, suggesting that (neuro)hormonal regulations may play an important role in controlling in vivo heart function.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Calcium-Transporting ATPases/metabolism , Isoproterenol/pharmacology , Myocardial Contraction/drug effects , Myocardium/metabolism , Animals , Calcium/pharmacology , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/genetics , Dobutamine/pharmacology , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Male , Mice , Mice, Transgenic , Myocardial Contraction/physiology , Phosphorylation , Receptors, Adrenergic, beta/metabolism , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases
SELECTION OF CITATIONS
SEARCH DETAIL
...