scholarly journals Parp-2 is required to maintain hematopoiesis following sublethal γ-irradiation in mice

Blood ◽  
2013 ◽  
Vol 122 (1) ◽  
pp. 44-54 ◽  
Author(s):  
Jordi Farrés ◽  
Juan Martín-Caballero ◽  
Carlos Martínez ◽  
Juan J. Lozano ◽  
Laura Llacuna ◽  
...  

Key Points Genetic inactivation of Parp-2 in mice, but not of Parp-1, resulted in bone marrow failure in response to sublethal γ-irradiation dose. Parp-2 plays an essential role in the DNA damage response in HSPC maintaining hematopoietic homeostasis under stress conditions.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4052-4052
Author(s):  
Travis Witt ◽  
Aloysius Klingelhutz ◽  
Erik Westin ◽  
Preeti Satyanarayana ◽  
Peter M. Lansdorp ◽  
...  

Abstract Dyskeratosis congenita (DC) is an inherited multisystem disorder of premature aging, typically characterized by bone marrow failure, mucosal leukoplakia, abnormal skin pigmentation, and nail dystrophy. The X-linked and autosomal dominant forms of DC are associated with mutations in genes that affect telomerase activity resulting in a decrease in telomere length. DC, like other bone marrow failure disorders, is associated with ineffective hematopoiesis and a cancer predisposition. Standard treatment of bone marrow failure or cancer requires cytotoxic therapy, and clinical observations suggest DC patients have an increased sensitivity to cytotoxic therapy. To explain this, we hypothesized that the short telomeres in somatic cells from DC patients could alter the activity and/or expression of several proteins involved in DNA repair or the response to cellular stress including p16, p53 and p21. Lymphocytes from five DC subjects and age-matched controls were stimulated to grow in vitro in the presence of various cytotoxic agents with different modes of action, including Taxol (antimitotic agent and microtubule inhibitor) and Etoposide (topoisomerase inhibitor and DNA damaging agent). In addition, we tested fibroblasts and keratinocyte extracted from skin biopsies from DC and control subjects that were serially passaged. Cellular proliferation and cell death were monitored by cell counts and flow cytometry. Western blotting was used to measure steady state and DNA damage- induced expression of tumor suppressor protein p53 and other proteins involved in DNA damage response signaling pathway, including p16 and p21 in relation to telomere length. Results of flow cytometry accompanied by direct visualization showed a decreased proliferation of DC lymphocytes compared to normal cells, and this growth disadvantage was further accentuated following cell exposure to cytotoxic agents. DC lymphocytes exposed to 10−6 M Taxol showed a decrease in cellular proliferation between 3 and 8 fold while normal control cells exposed to the same agents exhibited only a 3 to 4 fold decrease in cell growth. Similarly DC lymphocytes exposed to Etoposide were inhibited to a greater extent than control cells. Western blot analysis of whole cell lysates indicated a difference in DNA damage response proteins. Of note, lymphocytes from several DC subjects exposed to Taxol did not upregulate p53 expression, while inducible levels were noted in Taxol-treated control cells. In contrast, DC and control lymphocytes exposed to Etoposide upregulated p53 in a similar dose dependent manner. No differences were noted in DC versus control lymphocytes with regards to basal or chemotherapy induced p16 expression. Interestingly, late passage DC fibroblasts displayed enhanced basal expression of p16. These results support the clinical observation of increased “chemosensitivity” in DC subjects and suggest that diminished telomerase activity and premature telomere shortening may interfere with normal DNA damage and stress response pathways. These data are also consistent with our finding that DC fibroblasts, keratinocytes, and lymphocytes have a reduced cell proliferative lifespan. Further studies are needed to dissect the role of telomeres in the cellular response to various types of DNA damage.


2008 ◽  
Vol 29 (1) ◽  
pp. 229-240 ◽  
Author(s):  
Hua He ◽  
Yang Wang ◽  
Xiaolan Guo ◽  
Sonal Ramchandani ◽  
Jin Ma ◽  
...  

ABSTRACT The Protection of telomeres 1 (POT1) protein is a single-stranded telomere binding protein that is essential for proper maintenance of telomere length. Disruption of POT1 function leads to chromosome instability and loss of cellular viability. Here, we show that targeted deletion of the mouse Pot1b gene results in increased apoptosis in highly proliferative tissues. In the setting of telomerase haploinsufficiency, loss of Pot1b results in depletion of germ cells and complete bone marrow failure due to increased apoptosis, culminating in premature death. Pot1b −/ − mTR +/ − hematopoietic progenitor and stem cells display markedly reduced survival potential in vitro. Accelerated telomere shortening, increased G overhang and elevated number of chromosome end-to-end fusions that initiate an ATR-dependent DNA damage response were also observed. These results indicate an essential role for Pot1b in the maintenance of genome integrity and the long-term viability of proliferative tissues in the setting of telomerase deficiency. Interestingly, these phenotypes closely resemble those found in the human disease dyskeratosis congenita (DC), an inherited syndrome characterized by bone marrow failure, hyperpigmentation, and nail dystrophy. We anticipate that this mouse will serve as a useful model to further understand the pathophysiology of DC.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 799-799
Author(s):  
Dagmar Walter ◽  
Amelie Lier ◽  
Anja Geiselhart ◽  
Sina Huntscha ◽  
David Brocks ◽  
...  

Abstract Long-term quiescence has been proposed to preserve the genomic stability of hematopoietic stem cells (HSCs) during aging. The current models of HSC aging are limited in their ability to observe both DNA damage in vivo and the consequences of this damage upon hematopoiesis. Fanconi Anemia (FA) is a hereditary multisystem disorder, characterized by defective DNA damage response and progressive bone marrow failure in most patients. However, the existing genetic models of FA do not develop aplastic anemia, suggesting that cell-extrinsic factors may play a causal role. We sought to identify whether physiologic mediators of HSC activation could be used as agonists to provoke DNA damage and HSC attrition in vivo. Mice were treated with a range of agonists that promote the in vivo exit of HSC from a dormant state into active cycling (polyI:polyC; Interferon-α; G-CSF; TPO; and serial bleeding). Highly purified HSC demonstrated a rapid 3-5-fold induction of DNA damage after treatment with all agonists (p<0.01), as assessed by both enumerating γ-H2AX foci and by alkaline comet assay. Mechanistically, stress-induced exit from quiescence correlated with increased mitochondrial metabolism in HSC, as evaluated by elevated mitochondrial membrane potential (2-fold increased, p<0.01) and superoxide levels (1.5-fold increased, p<0.05). Critically, we could directly implicate these reactive oxygen species in DNA damage as we observed a 1.4-fold increase in 8-Oxo-dG lesions in HSC that had been activated into cycle in vivo(p<0.05). At 48 h post-treatment, γ-H2AX levels began to decrease and this repair was concomitant with an induction of the FA signaling pathway in HSC, as demonstrated by both increased levels of FA gene expression and elevated FANCD2 foci (4-fold induction, p<0.01). Treatment of Fanca-/- mice with polyI:polyC led to a HSC proliferative response comparable to wild type (WT) mice but resulted in a 2-fold higher level of activation-induced DNA damage (p<0.05), demonstrating that this repair pathway is involved in resolving activation-induced DNA damage. Four rounds of serial in vivo activation led to a permanent depletion of the most primitive label-retaining Fanca-/- HSC and this correlated with a 4-fold depletion of functional HSC (p<0.01) as defined by competitive repopulation assays. Subsequent rounds of HSC activation with polyI:polyC resulted in the onset of a severe aplastic anemia (SAA) in 33% of treated Fanca-/- mice but not in any of the WT controls. SSA was characterized by a dramatic reduction in bone marrow (BM) cellularity, profound thrombocytopenia (21-246x106 platelets/ml), leukocytopenia (0.4-0.5x106 WBC/ml), neutropenia (0.03-0.1x106/ml) and anemia (1.5-2.3 g/dL Hb). Examination of BM HSC/progenitors demonstrated nearly complete loss of HSC, MPP, CMP and CLP (depletion of ≥33x, 8x, 4x and 12x respectively compared to PBS-treated Fanca-/-controls). Taken together, these data demonstrates that enforced exit from dormancy in vivo leads to de novo DNA damage in HSC, which is repaired by activation of a FA-dependent DNA damage response. Furthermore, the highly penetrant bone marrow failure observed in Fanconi anemia patients can be recapitulated by the serial application of a physiologic HSC activating signal to Fanca-/- mice. This suggests that the BM failure in FA may be caused by an aberrant response to HSC activation, most likely during exposure to infection or other physiologic stressors. These data provides a novel link between pro-inflammatory cytokines, DNA damage and HSC dysfunction and may have important clinical implications relevant to both prevention of BM failure in FA and in the study of age-related hematopoietic defects in non-FA patients. Moreover, these data provide the first evidence that FA knockout mouse models accurately recapitulate and provide novel insights into the etiology of BM failure in patients with FA. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (24) ◽  
pp. 2631-2641 ◽  
Author(s):  
Brenton G. Mar ◽  
S. Haihua Chu ◽  
Josephine D. Kahn ◽  
Andrei V. Krivtsov ◽  
Richard Koche ◽  
...  

Key Points Alterations of SETD2, a histone 3 lysine 36 trimethyl (H3K36me3) transferase leads to resistance to DNA damaging-chemotherapy in leukemia. Low H3K36me3 levels impair DNA damage response and increase mutation rate, which may be targeted by H3K36me3 demethylase inhibition.


2018 ◽  
Vol 2 (6) ◽  
pp. 586-596 ◽  
Author(s):  
Manuela Germeshausen ◽  
Phil Ancliff ◽  
Jaime Estrada ◽  
Markus Metzler ◽  
Eva Ponstingl ◽  
...  

Key Points Germ line mutations in MECOM cause a heterogeneous bone marrow failure syndrome with congenital hypomegakaryocytic thrombocytopenia. MECOM-associated syndrome includes various organ malformations with variable penetrance, including radioulnar synostosis.


Blood ◽  
2013 ◽  
Vol 122 (14) ◽  
pp. 2453-2459 ◽  
Author(s):  
Andres Jerez ◽  
Michael J. Clemente ◽  
Hideki Makishima ◽  
Hanna Rajala ◽  
Ines Gómez-Seguí ◽  
...  

Key PointsSTAT3+ T cells are found not only in detected concomitant LGL-BMFs, but in cases in which an LGL expansion was not suspected. Transformation via acquisition of a somatic mutation in T cells may be a mechanism of immune, mainly hypoplastic, bone marrow failure.


Blood ◽  
2013 ◽  
Vol 121 (25) ◽  
pp. 5078-5087 ◽  
Author(s):  
Polina Stepensky ◽  
Ann Saada ◽  
Marianne Cowan ◽  
Adi Tabib ◽  
Ute Fischer ◽  
...  

Key Points VPS45 is a new gene associated with severe infections and bone marrow failure in infancy that can be treated by bone marrow transplantation. The mutation affects intracellular storage and transport and results in increased programmed cell death in neutrophils and bone marrow.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 723-723
Author(s):  
Alexandra Sobeck ◽  
Stacie Stone ◽  
Bendert deGraaf ◽  
Vincenzo Costanzo ◽  
Johan deWinter ◽  
...  

Abstract Fanconi anemia (FA) is a genetic disorder characterized by hypersensitivity to DNA crosslinking agents and diverse clinical symptoms, including developmental anomalies, progressive bone marrow failure, and predisposition to leukemias and other cancers. FA is genetically heterogeneous, resulting from mutations in any of at least eleven different genes. The FA proteins function together in a pathway composed of a mulitprotein core complex that is required to trigger the DNA-damage dependent activation of the downstream FA protein, FANCD2. This activation is thought to be the key step in a DNA damage response that functionally links FA proteins to major breast cancer susceptibility proteins BRCA1 and BRCA2 (BRCA2 is FA gene FANCD1). The essential function of the FA proteins is unknown, but current models suggest that FA proteins function at the interface between cell cycle checkpoints, DNA repair and DNA replication, and are likely to play roles in the DNA damage response during S phase. To provide a platform for dissecting the key functional events during S-phase, we developed cell-free assays for FA proteins based on replicating extracts from Xenopus eggs. We identified the Xenopus homologs of human FANCD2 (xFANCD2) and several of the FA core complex proteins (xCCPs), and biochemically characterized these proteins in replicating cell-free extracts. We found that xCCPs and a modified isoform of xFANCD2 become associated with chromatin during normal and disrupted DNA replication. Blocking initiation of replication with geminin demonstrated that association of xCCPs and xFANCD2 with chromatin occurs in a strictly replication-dependent manner that is enhanced following DNA damage by crosslinking agents or by addition of aphidicolin, an inhibitor of replicative DNA polymerases. In addition, chromatin binding of xFANCD2, but not xBRCA2, is abrogated when xFANCA is quantitatively depleted from replicating extracts suggesting that xFANCA promotes the loading of xFANCD2 on chromatin. The chromatin-association of xFANCD2 and xCCPs is diminished in the presence of caffeine, an inhibitor of checkpoint kinases. Taken together, our data suggest a model in which the ordered loading of FA proteins on chromatin is required for processing a subset of DNA replication-blocking lesions that are resolved during late stages of replication.


Sign in / Sign up

Export Citation Format

Share Document