Telomere shortening during aging of human osteoblasts in vitro and leukocytes in vivo: lack of excessive telomere loss in osteoporotic patients

1999 ◽  
Vol 106 (3) ◽  
pp. 261-271 ◽  
Author(s):  
Marie Kveiborg ◽  
Moustapha Kassem ◽  
Bente Langdahl ◽  
Erik Fink Eriksen ◽  
Brian F.C Clark ◽  
...  
Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1620-1620
Author(s):  
Angela Gueli ◽  
Marco Ruella ◽  
Alessandra Risso ◽  
Tiziana Spatola ◽  
Hui Jing Hu ◽  
...  

Abstract Abstract 1620 Introduction Patients with lymphoproliferative disease very often require treatments with chemotherapy. Several recent reports suggest that the exposure of hematopoietic cells to DNA-damaging substances, such as chemotherapeutic drugs, may trigger the ageing process and induce premature cell ageing. A good indicator of cell replication history and thus of cell ageing is the length of telomeres. Indeed, telomere shortening and/or telomere dysfunction have been documented in patients receiving chemotherapy. The loss of telomere sequences has been linked to the increased risk of developing secondary malignancy in subjects previously treated with chemotherapy. However, it is still unexplained whether telomere loss is secondary to direct damage of bone marrow (BM) cells by cytotoxic drugs, or merely the consequence of increased BM cell proliferation after exposure to chemotherapy. Moreover, the time course for the onset of chemotherapy-induced telomere shortening and the cell types mainly involved in the premature cell aging induced by chemotherapy remain to be elucidated. In the present study changes in telomere length (TL) following chemotherapy were evaluated both in vivo and in vitro. Main aims of the study were: i. to define whether TL shortening following chemotherapy is reversible or permanent; ii. to verify whether TL shortening is a phenomenon induced by extensive chemotherapy treatments or it may occur even after minimal drug exposures; iii. to identify possible cell populations that are particularly susceptible to drug-induced telomere loss. Patients and methods Mononuclear Cells (MNC) or granulocytes were obtained from BM and/or Peripheral blood (PB) cells from 31 lymphoma patients treated with high-dose chemotherapy and autograft and from 14 lymphoma patients undergoing chemotherapy (6 CHOP, 1 CVP, 1 MINE, 1 DHAP and 5 ABVD). Overall, 27 patients of the autograft and 13 patients of the conventional chemotherapy groups were at their first treatment line. Median age of patients was 45 years. TL was assessed on granulocytes and MNC by Southern Blotting, as previously reported (Ricca et al, Leukemia 2005). In vitro studies were performed on cultured mesenchymal stem cells (MSCs). Briefly, MNC were obtained from BM of normal volunteers or patients undergoing routine diagnostic procedures; cells were seeded in MEM-alpha medium and 10% platelet lysates and fed at 3–4 day intervals; cultured MSC were identified for positivity of CD105, CD90, CD29, CD44 and then assayed for response to DNA-damaging drugs between the second and the third passage in culture. Two chemotherapeutic drugs were employed, Doxorubicin (Doxo) and etoposide (Eto). Cells were incubated for 2 hours with decreasing doses of the tested drugs, and 10 nM Doxo and 500 ng/ml Eto were the highest doses of the drugs that were used without any distress on cell proliferation and cell viability. The 2-hour exposure to chemotherapy was repeated at 7 day intervals up to four times. TL was evaluated both by flow-fish and southern-blot analysis. Results A marked reduction in TL was detected in all patients undergoing autograft in PB granulocytes and in BM cells, compared to age-matched controls; the degree of TL loss remained detectable even in patients up to 10 yrs. since autograft. For patients receiving conventional chemotherapy, a significant TL shortening could be detected in granulocyte obtained after chemotherapy compared to pre-treatment values (p=0.029), while no significant variations could be documented in MNC. TL shortening was detectable already after the first chemotherapy course in six patients and at the second in four patients. A marked TL shortening occurred in cultured MSC after exposure to sub-lethal doses of Doxo and Eto. Initial TL shortening was detectable already at 5 days after drug exposure, with progressive reduction compared with untreated cells at 7, 14, 21, and 28 days in culture. Following a single exposure, MSCs were unable to regain the lost telomere sequences for up to 28 days in culture. Conclusions The results indicate that TL shortening: i. is a permanent signature of the previous DNA damage in BM cells exposed to chemotherapy; ii. is a phenomenon that can be detected early following chemotherapy exposure, even with low drug dosages; iii. can be most easily detectable in myeloid cells, in particular in granulocytes, although also BM mesenchymal cells may be susceptible to drug-induced telomere loss. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Jonathan M. Lawton ◽  
Mariam Habib ◽  
Bingkui Ma ◽  
Roger A. Brooks ◽  
Serena M. Best ◽  
...  

2018 ◽  
Vol 373 (1741) ◽  
pp. 20160446 ◽  
Author(s):  
Pat Monaghan ◽  
Susan E. Ozanne

Much telomere loss takes place during the period of most rapid growth when cell proliferation and potentially energy expenditure are high. Fast growth is linked to reduced longevity. Therefore, the effects of somatic cell proliferation on telomere loss and cell senescence might play a significant role in driving the growth-lifespan trade-off. While different species will have evolved a growth strategy that maximizes lifetime fitness, environmental conditions encountered during periods of growth will influence individual optima. In this review, we first discuss the routes by which altered cellular conditions could influence telomere loss in vertebrates, with a focus on oxidative stress in both in vitro and in vivo studies. We discuss the relationship between body growth and telomere length, and evaluate the empirical evidence that this relationship is generally negative. We further discuss the potentially conflicting hypotheses that arise when other factors are taken into account, and the further work that needs to be undertaken to disentangle confounding variables. This article is part of the theme issue ‘Understanding diversity in telomere dynamics’.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3857-3857
Author(s):  
Dominik G.F. Wolf ◽  
Anna M. Wolf ◽  
Christian Koppelstaetter ◽  
Holger F. Rumpold ◽  
Gert Mayer ◽  
...  

Abstract The expandability of CD4+CD25+ regulatory T-cells (Treg) has been shown in vitro and in vivo. Activation of telomerase activity is a prerequisite for clonal expansion and telomere maintenance in T-cells. There is currently no data available on the expression and function of telomerase in proliferating Treg. Analyses of telomere length by flow-FISH, real-time PCR and Southern blotting revealed that Treg isolated from healthy human volunteers have significantly shortened telomeres when compared to CD4+CD25− T-cells. However, telomere length is not further shortened in Treg isolated from the peripheral blood of cancer patients, despite the observation that the regulatory T-cell pool of these patients was significantly enlarged. To gain further insight into maintenance of telomere length of Treg, we induced in vitro proliferation of Treg by stimulation with anti-CD3 and IL-2. This led to a rapid increase of telomerase activity, as determined by PCR-ELISA. However, when we focused on the proliferating fraction of Treg using a sorting strategy based on the dilution of CFSE, we could show a significant telomere shortening in Treg with high proliferative and immmuno-suppressive capacity. Of note, proliferating CFSElow Treg are characterized by high telomerase activity, which however seems to be insufficient to avoid further telomere shortening under conditions of strong in vitro stimulation. In contrast, under conditions of in vivo expansion of Treg in cancer patients, the induction of telomerase activity is likely to compensate for further telomere erosion. These data might be of importance when considering the application of in vitro expanded Treg for the treatment of GvHD or autoimmune diseases, as telomere shortening might be associated with genomic instability.


2010 ◽  
Vol 108 (1) ◽  
pp. 73-78 ◽  
Author(s):  
Catherine Cifuentes-Rojas ◽  
Kalpana Kannan ◽  
Lin Tseng ◽  
Dorothy E. Shippen

Telomerase is a ribonucleoprotein (RNP) reverse transcriptase whose essential RNA subunit (TER) functions as a template for telomere repeat synthesis. Here we report the identification of two divergent TER moieties in the flowering plant Arabidopsis thaliana. Although both TER1 and TER2 copurify with telomerase activity and serve as templates for telomerase in vitro, depletion of TER1, but not TER2, leads to decreased telomerase activity and progressive telomere shortening in vivo. Moreover, mutation of the templating domain in TER1 results in the incorporation of mutant telomere repeats on chromosome ends. Thus, TER1 provides the major template for telomerase in vivo. We also show that POT1a binds TER1 with a Kd of 2 × 10-7 M and the two components assemble into an enzymatically active RNP in vivo. In contrast, TER1-POT1b and TER2-POT1a associations were not observed. In other organisms POT1 proteins bind telomeric DNA and provide chromosome end protection. We propose that duplication of TER and POT1 in Arabidopsis fueled the evolution of novel protein–nucleic acid interactions and the migration of POT1 from the telomere to the telomerase RNP.


2020 ◽  
Author(s):  
Yun Gong ◽  
Junxiao Yang ◽  
Xiaohua Li ◽  
Cui Zhou ◽  
Yu Chen ◽  
...  

AbstractOsteoblasts are multifunctional bone cells, which play essential roles in bone formation, angiogenesis regulation, as well as maintenance of hematopoiesis. Although both in vivo and in vitro studies on mice have identified several potential osteoblast subtypes based on their different transition stages or biological responses to external stimuli, the categorization of primary osteoblast subtypes in vivo in humans has not yet been achieved. Here, we used single-cell RNA sequencing (scRNA-seq) to perform a systematic cellular taxonomy dissection of freshly isolated human osteoblasts. Based on the gene expression patterns and cell lineage reconstruction, we identified three distinct cell clusters including preosteoblasts, mature osteoblasts, and an undetermined rare osteoblast subpopulation. This novel subtype was mainly characterized by the nuclear receptor subfamily 4 group A member 1 and 2 (NR4A1 and NR4A2), and its existence was confirmed by immunofluorescence staining. Trajectory inference analysis suggested that the undetermined cluster, together with the preosteoblasts, are involved in the regulation of osteoblastogenesis and also give rise to mature osteoblasts. Investigation of the biological processes and signaling pathways enriched in each subpopulation revealed that in addition to bone formation, preosteoblasts and undetermined osteoblasts may also regulate both angiogenesis and hemopoiesis. Finally, we demonstrated that there are systematic differences between the transcriptional profiles of human osteoblasts in vivo and mouse osteoblasts both in vivo and in vitro, highlighting the necessity for studying bone physiological processes in humans rather than solely relying on mouse models. Our findings provide novel insights into the cellular heterogeneity and potential biological functions of human primary osteoblasts at the single-cell level, which is an important and necessary step to further dissect the biological roles of osteoblasts in bone metabolism under various (patho-) physiological conditions.


2010 ◽  
Vol 10 ◽  
pp. 727-741 ◽  
Author(s):  
Raya Saab

Cellular senescence is a tumor-suppressor mechanism that has been shown to occur in response to multiple signals, including oncogenic stress, DNA damage, oxidative stress, telomere shortening, and other tumor-promoting insults. Over the past decade, much has been uncovered regarding the phenotype of this tumor-suppressor response and the underlying pathways necessary for its establishment. However, we have also learned that the intricate details of signaling pathways underlying senescence as a tumor-suppressor response are very much context dependent. In addition, cross-talk among pathways, and negative and positive feedback loops, all complicate our understanding of this process. This short review attempts to summarize what is known to date regarding senescence in tumor suppression, bothin vitroandin vivo. Further insights into pathways necessary for senescence will hopefully identify appropriate targets for interventions to not only induce senescence as a treatment of cancerous lesions, but also to maintain this state in premalignant lesions in an effort to prevent progression to cancer.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 639-639
Author(s):  
Masood A. Shammas ◽  
Hemanta Koley ◽  
Pierfrancesco Tassone ◽  
Paola Neri ◽  
Alexei Protopopov ◽  
...  

Abstract Telomerase activity is either low or completely absent in most normal somatic cells; while it is elevated in most cancer cells providing unlimited proliferative potential by preventing telomere shortening. The inhibitors of telomerase, therefore, induce telomere shortening leading to apoptotic cell death in tumor cells while having little or no effect on normal diploid cells. We have evaluated the in vitro and in vivo efficacy of thio-phosphoramidate oligonucleotide specifically targeting the RNA component of telomerase (GRN163L) with demonstrated nuclear uptake by >99% cells without the transfection enhancer. Delivery of GRN163L (1 μM) to MM cells (INA6 and ARP) was specifically associated with complete loss of telomerase activity as early as 6 hrs following exposure and was accompanied by a reduction in myeloma cell growth and survival. Treatment of INA6 cells with GRN163L for three weeks induced 96±4% and 100% cell death at 0.5 and 1 μM concentrations, respectively. ARP cells, which express higher levels of telomerase activity and have longer telomeres, showed 67±4% cell death at 5 weeks with 0.5 μM inhibitor and 82±3% and 100% cell death at 4 and 5 weeks, respectively, with 2 μM GRN163L. The apoptotic cell death was confirmed in 51% INA6 cells at two weeks and in >80% ARP cells at four weeks. Apoptosis was associated with reduction in mean Telomere Fluorescence Intensity (TFI) on interphase chromosomes from 87.1±6.2 in control oligo treated INA6 cells to 36.2±2 (2.4 fold) in GRN163L treated cells. Moreover, GRN163L treatment was also associated with a similar reduction in number of chromosomes with detectable telomeres, indicating development of telomere-free ends. We have confirmed in vivo efficacy of GRN163L in a SCID-hu murine model of multiple myeloma. Following growth of GFP-transduced myeloma cells in the fetal bone chip introduced into the mice, GRN163L was injected on alternate days. In two independent experiments significant reduction in tumor cell growth, as measured by reduction in human myeloma related protein, and better survival than mice injected with control oligo was observed. We have now evaluated efficacy of combination of GRN163L with other novel agents. We have observed synergistic activity with Hsp90 inhibitor 17AAG on myeloma cell death. Addition of 17AAG (0.05 μM) to myeloma cells pre-treated with GRN163L (1 μM) for one week led to complete growth arrest within four days compared to continued growth of cells not pre-treated with GRN-163. These data provide the preclinical rationale for clinical evaluation of GRN163L in myeloma and in combination with Hsp90 inhibitor.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1922-1922
Author(s):  
Mark Zijlmans ◽  
Susan Swiggers ◽  
Maria Rife Soler ◽  
Berna Beverloo

Abstract Immortal cell growth is considered the hallmark of tumor cells. In contrast, normal cells have a limited proliferative capacity of 40–60 cell divisions, also known as the Hayflick limit. The limited proliferative capacity of normal cells relates to gradual telomere shortening as a consequence of the end-replication problem. Upon critical telomere shortening, cells enter a non-replicative but viable state referred to as replicative senescence. These replicative senescent cells stain blue in a beta-Galactosidase assay and activate DNA double-strand break repair pathways at telomeres (e.g. gamma-H2AX foci). In human fibroblast models, escape from senescence results from loss of p53 and Rb function. Escape is associated with reactivation of telomerase. High levels of telomerase, as observed in germ cells and most tumor cells, allow for immortal cell growth. Recently, we demonstrated low levels of telomerase in AML patients with t(8;21) or inv(16) (Swiggers et al, G.C.C. 2006). Interestingly, levels of telomerase in these AML samples were similar to levels of telomerase in normal bone marrow progenitor cells. We hypothesized that AML without re-activated telomerase may still have intact senescence pathways that limit the proliferative capacity of normal cells. This hypothesis was addressed by studying AML patient samples without telomerase re-activation, i.e., t(8;21), t(15;17) or inv(16) (n=10), and a control group of AML with telomerase re-activation (multiple gains/losses of genetic material, n=8). AML samples werelong-time cultured in vitro in the presence of hematopoietic growth factors (range 3–6 weeks),analyzed in vivo following transplantation in NOD-SCID mice andin patients at time of relapse. Cells with all characteristics of replicative senescence, i.e. enlarged, viable, non-proliferating, blue-coloring in beta-Galactosidase assay, critical short telomeres and gamma-H2AX foci at telomeres, were clearly observed in all AML samples with t(8;21), t(15;17) or inv(16). Gradual telomere shortening was observed in these AML cells in vitro upon long-term culture, in vivo after transplantation in NOD-SCID mice and in vivo in patients at relapse compared to time of diagnosis, indicating that these AML cells do not have an adequate telomere maintenance mechanism. We conclude that AML cells with t(8;21), t(15;17) or inv(16) are characterized by intact pathways that induce replicative senescence. Intact pathways that limit proliferative lifespan may be critical to the high cure rates following chemotherapy treatment of patients with good-risk AML.


Sign in / Sign up

Export Citation Format

Share Document