Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cell ; 164(4): 668-80, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26871632

ABSTRACT

Mouse embryonic stem cells (ESCs) are maintained in a naive ground state of pluripotency in the presence of MEK and GSK3 inhibitors. Here, we show that ground-state ESCs express low Myc levels. Deletion of both c-myc and N-myc (dKO) or pharmacological inhibition of Myc activity strongly decreases transcription, splicing, and protein synthesis, leading to proliferation arrest. This process is reversible and occurs without affecting pluripotency, suggesting that Myc-depleted stem cells enter a state of dormancy similar to embryonic diapause. Indeed, c-Myc is depleted in diapaused blastocysts, and the differential expression signatures of dKO ESCs and diapaused epiblasts are remarkably similar. Following Myc inhibition, pre-implantation blastocysts enter biosynthetic dormancy but can progress through their normal developmental program after transfer into pseudo-pregnant recipients. Our study shows that Myc controls the biosynthetic machinery of stem cells without affecting their potency, thus regulating their entry and exit from the dormant state.


Subject(s)
Embryonic Stem Cells/cytology , Genes, myc , Proto-Oncogene Proteins c-myc/genetics , Animals , Blastocyst/metabolism , Cell Proliferation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Female , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL
2.
Haematologica ; 102(3): 445-453, 2017 03.
Article in English | MEDLINE | ID: mdl-27742772

ABSTRACT

In the bone marrow, endothelial cells are a major component of the hematopoietic stem cell vascular niche and are a first line of defense against inflammatory stress and infection. The primary response of an organism to infection involves the synthesis of immune-modulatory cytokines, including interferon alpha. In the bone marrow, interferon alpha induces rapid cell cycle entry of hematopoietic stem cells in vivo However, the effect of interferon alpha on bone marrow endothelial cells has not been described. Here, we demonstrate that acute interferon alpha treatment leads to rapid stimulation of bone marrow endothelial cells in vivo, resulting in increased bone marrow vascularity and vascular leakage. We find that activation of bone marrow endothelial cells involves the expression of key inflammatory and endothelial cell-stimulatory markers. This interferon alpha-mediated activation of bone marrow endothelial cells is dependent in part on vascular endothelial growth factor signaling in bone marrow hematopoietic cell types, including hematopoietic stem cells. Thus, this implies a role for hematopoietic stem cells in remodeling of the bone marrow niche in vivo following inflammatory stress. These data increase our current understanding of the relationship between hematopoietic stem cells and the bone marrow niche under inflammatory stress and also clarify the response of bone marrow niche endothelial cells to acute interferon alpha treatment in vivo.


Subject(s)
Bone Marrow/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Interferon-alpha/metabolism , Stem Cell Niche , Animals , Biomarkers , Bone Marrow/blood supply , Bone Marrow Cells/metabolism , Capillary Permeability , Cell Proliferation , Cellular Microenvironment , Female , Hematopoiesis , Hematopoietic Stem Cells/cytology , Immunophenotyping , Interferon-alpha/genetics , Interferon-alpha/pharmacology , Mice , Mice, Knockout , Models, Biological , Neovascularization, Physiologic , Phenotype , Poly I-C/pharmacology , Signal Transduction/drug effects , Stress, Physiological , Vascular Endothelial Growth Factor A/biosynthesis
3.
Subcell Biochem ; 50: 189-209, 2010.
Article in English | MEDLINE | ID: mdl-20012583

ABSTRACT

Genomic DNA is constantly damaged by exposure to exogenous and endogenous agents. Bulky adducts such as UV-induced cyclobutane pyrimidine dimers (CPDs) in the template DNA present a barrier to DNA synthesis by the major eukaryotic replicative polymerases including DNA polymerase delta. Translesion synthesis (TLS) carried out by specialized DNA polymerases is an evolutionarily conserved mechanism of DNA damage tolerance. The Y family of DNA polymerases, including DNA polymerase eta (Pol eta), the subject of this chapter, play a key role in TLS. Mutations in the human POLH gene encoding Pol eta underlie the genetic disease xeroderma pigmentosum variant (XPV), characterized by sun sensitivity, elevated incidence of skin cancer, and at the cellular level, by delayed replication and hypermutability after UV-irradiation. Pol eta is a low fidelity enzyme when copying undamaged DNA, but can carry out error-free TLS at sites of UV-induced dithymine CPDs. The active site of Pol eta has an open conformation that can accommodate CPDs, as well as cisplatin-induced intrastrand DNA crosslinks. Pol eta is recruited to sites of replication arrest in a tightly regulated process through interaction with PCNA. Pol eta-deficient cells show strong activation of downstream DNA damage responses including ATR signaling, and accumulate strand breaks as a result of replication fork collapse. Thus, Pol eta plays an important role in preventing genome instability after UV- and cisplatin-induced DNA damage. Inhibition of DNA damage tolerance pathways in tumors might also represent an approach to potentiate the effects of DNA damaging agents such as cisplatin.


Subject(s)
DNA Damage , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Humans
4.
Stem Cell Res Ther ; 6: 136, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26204937

ABSTRACT

INTRODUCTION: Bone marrow-derived stromal cells (BMSCs), also known as mesenchymal stem cells, are the focus of intensive efforts worldwide to elucidate their function and biology. Despite the importance of BMSC migration for their potential therapeutic uses, the mechanisms and signalling governing stem cell migration are still not fully elucidated. METHODS: We investigated and detailed the effects of MCP-1 activation on BMSCs by using inhibitors of G protein-coupled receptor alpha beta (GPCR αß), ROCK (Rho-associated, coiled-coil containing protein kinase), and PI3 kinase (PI3K). The effects of MCP-1 stimulation on intracellular signalling cascades were characterised by using immunoblotting and immunofluorescence. The effectors of MCP-1-mediated migration were investigated by using migration assays (both two-dimensional and three-dimensional) in combination with inhibitors. RESULTS: We established the kinetics of the MCP-1-activated signalling cascade and show that this cascade correlates with cell surface re-localisation of chemokine (C motif) receptor 2 (CCR2) (the MCP-1 receptor) to the cell periphery following MCP-1 stimulation. We show that MCP-1-initiated signalling is dependent on the activation of ßγ subunits from the GPCR αßγ complex. In addition, we characterise a novel role for PI3Kγ signalling for the activation of both PAK and ERK following MCP-1 stimulation. We present evidence that the Gßγ complex is responsible for PI3K/Akt, PAK, and ERK signalling induced by MCP-1 in BMSCs. Importantly, we found that, in BMSCs, inhibition of ROCK significantly inhibits MCP-1-induced chemotactic migration, in contrast to previous reports in other systems. CONCLUSIONS: Our results indicate differential chemotactic signalling in mouse BMSCs, which has important implications for the translation of in vivo mouse model findings into human trials. We identified novel components and interactions activated by MCP-1-mediated signalling, which are important for stem cell migration. This work has identified additional potential therapeutic targets that could be manipulated to improve BMSC delivery and homing.


Subject(s)
Chemotaxis , GTP-Binding Protein beta Subunits/metabolism , Mesenchymal Stem Cells/physiology , rho-Associated Kinases/metabolism , Animals , Antigens/metabolism , Cells, Cultured , Chemokine CCL2/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
5.
Cell Stem Cell ; 17(4): 422-34, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26299573

ABSTRACT

Infections are associated with extensive platelet consumption, representing a high risk for health. However, the mechanism coordinating the rapid regeneration of the platelet pool during such stress conditions remains unclear. Here, we report that the phenotypic hematopoietic stem cell (HSC) compartment contains stem-like megakaryocyte-committed progenitors (SL-MkPs), a cell population that shares many features with multipotent HSCs and serves as a lineage-restricted emergency pool for inflammatory insults. During homeostasis, SL-MkPs are maintained in a primed but quiescent state, thus contributing little to steady-state megakaryopoiesis. Even though lineage-specific megakaryocyte transcripts are expressed, protein synthesis is suppressed. In response to acute inflammation, SL-MkPs become activated, resulting in megakaryocyte protein production from pre-existing transcripts and a maturation of SL-MkPs and other megakaryocyte progenitors. This results in an efficient replenishment of platelets that are lost during inflammatory insult. Thus, our study reveals an emergency machinery that counteracts life-threatening platelet depletions during acute inflammation.


Subject(s)
Blood Platelets/pathology , Inflammation/pathology , Megakaryocyte Progenitor Cells/pathology , Thrombopoiesis , Animals , Blood Platelets/physiology , Cell Lineage , Cell Proliferation , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Megakaryocyte Progenitor Cells/physiology , Mice
6.
Ann N Y Acad Sci ; 1310: 51-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24611873

ABSTRACT

The immune response to infection is a rapid and multifaceted process. Infection affects homeostasis within the hematopoietic stem cell (HSC) niche, as lost immune cells must be replaced by HSCs. During the immune response, interferon is produced. Surprisingly, HSCs respond directly to interferon, entering the cell cycle from even the most dormant state. The complex response of both the HSCs and the niche to infection is a unique platform on which to consider HSC-niche interactions. Here, we comment on the contribution of the immune system to the niche and on the direct and indirect effect that infection has on HSCs in the niche.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunity, Innate/physiology , Infections/immunology , Stem Cell Niche/immunology , Animals , Humans , Mice
7.
Int J Hematol ; 96(5): 649-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23076878

ABSTRACT

Anthracyclines, including doxorubicin, are widely used in the treatment of leukemia. While the effects of doxorubicin on hematopoietic cells have been characterized, less is known about the response of human mesenchymal stem cells (hMSCs) in the bone marrow stroma to anthracyclines. We characterized the effect of doxorubicin on key DNA damage responses in hMSCs, and compared doxorubicin sensitivity and DNA damage response activation between isolated hMSCs and the chronic myelogenous leukemia cell line, K562. Phosphorylation of H2AX, Chk1, and RPA2 was more strongly activated in K562 cells than in hMSCs, at equivalent doses of doxorubicin. hMSCs were relatively resistant to doxorubicin such that, following exposure to 15 µM doxorubicin, the level of cleaved caspase-3 detected by western blotting was lower in hMSCs compared to K562 cells. Flow cytometric analysis of cell cycle progression demonstrated that exposure to doxorubicin induced G2/M phase arrest in hMSCs, while 48 h after exposure, 15.6 % of cells were apoptotic, as determined from the percentage of cells having sub-G1 DNA content. We also show that the doxorubicin sensitivity of hMSCs isolated from a healthy donor was comparable to that of hMSCs isolated from a chronic lymphocytic leukemia patient. Overall, our results demonstrate that high doses of doxorubicin induce the DNA damage response in hMSCs, and that cultured hMSCs are relatively resistant to doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , DNA Damage , Doxorubicin/adverse effects , Mesenchymal Stem Cells/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Checkpoint Kinase 1 , Doxorubicin/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Histones/metabolism , Humans , Inorganic Pyrophosphatase/metabolism , K562 Cells , M Phase Cell Cycle Checkpoints/drug effects , Male , Mesenchymal Stem Cells/pathology , Mitochondrial Proteins/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism
8.
Cell Cycle ; 10(21): 3768-77, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22037398

ABSTRACT

DNA damaging agents are widely used in treatment of hematogical malignancies and solid tumors. While effects on hematopoietic stem cells have been characterized, less is known about the DNA damage response in human mesenchymal stem cells (hMSCs) in the bone marrow stroma, progenitors of osteoblasts, chondrocytes and adipocytes. To elucidate the response of undifferentiated hMSCs to γ-irradiation and cisplatin, key DNA damage responses have been characterised in hMSCs from normal adult donors. Cisplatin and γ-irradiation activated the DNA damage response in hMSCs, including induction of p53 and p21, and activation of PI3 kinase-related protein kinase (PIKK)-dependent phosphorylation of histone H2AX on serine 139, and replication protein A2 on serine4/serine8. Chemical inhibition of ATM or DNA-PK reduced DNA damage-induced phosphorylation of H2AX, indicating a role for both PIKKs in the response of hMSCs to DNA damage. Consistent with repair of DNA strand breaks, γ-H2AX staining decreased by 24 hours following gamma-irradiation. γ-Irradiation arrested hMSCs in the G 1 phase of the cell cycle, while cisplatin induced S-phase arrest, mediated in part by the ATR/Chk1 checkpoint pathway. In hMSCs isolated from a chronic lymphocytic leukemia (CLL) patient, p53 and p21 were induced by cisplatin and γ-irradiation, while RPA2 was phosphorylated on serine4/8 in particular following cisplatin. Compared to peripheral blood lymphocytes or the leukemia cell line K562, both normal hMSCs and CLL-derived hMSCs were more resistant to cisplatin and γ-irradiation. These results provide insights into key pathways mediating the response of bone marrow-derived hMSCs to DNA damaging agents used in cancer treatment.


Subject(s)
Cisplatin/pharmacology , DNA Damage , Gamma Rays , Mesenchymal Stem Cells/drug effects , Mutagens/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Adducts , DNA Breaks/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Histones/metabolism , Humans , Mesenchymal Stem Cells/radiation effects , Phosphorylation , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
9.
Curr Cancer Drug Targets ; 10(4): 411-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20370690

ABSTRACT

Human mesenchymal stem cells (hMSCs) consist of cells that can differentiate into mesenchymal tissues, including osteoblasts, adipocytes and chondrocytes. hMSCs constitute a particular stem cell niche in the stromal compartment of the bone marrow, and also play a role in maintaining the normal function of haematopoietic stem cells. Furthermore, hMSCs localise to solid tumours, and can modulate cancer cell function through secretion of paracrine signals. While hMSCs, either in the bone marrow, or in the microenvironment of a tumour, will be targeted by DNA damaging agents used in cancer therapy, the response of the hMSC population to DNA damage is not well understood. In their role as progenitor cells, genomic DNA damage to hMSCs during cancer therapy could generate a population of surviving cells that can go on to give rise to secondary tumours. A better understanding of the response of hMSCs to DNA damage could provide new insights into the effects of cancer treatments, as well as into the development of treatment-associated secondary cancers. The article will review the relationship of hMSCs to cancer, with a focus on the response of hMSCs to DNA damaging agents.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Mesenchymal Stem Cells/drug effects , Neoplasms/drug therapy , Humans , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL