Iron Deprivation Impairs Proliferation of CALM-AF10 leukemia Cells in Vitro and in Vivo

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1386-1386
Author(s):  
Jessica L Heath ◽  
Joshua Weiss ◽  
Paula B. Scotland ◽  
Catherine P. Lavau ◽  
Daniel S. Wechsler

Abstract Abstract 1386 Background: The CALM-AF10 fusion protein arises from the t(10;11) chromosomal translocation and is found in 8–10% of T-cell acute lymphoblastic leukemias and a smaller percentage of acute myeloid leukemias; these hematopoietic malignancies are associated with a poor clinical outcome. The precise contributions of the Clathrin Assembly Lymphoid Myeloid leukemia (CALM) protein to leukemogenesis remain uncertain. CALM plays a role in clathrin-dependent endocytosis, which mediates the entry of growth factor receptors and nutrients into cells and is essential for the internalization of iron-bound transferrin. We have previously shown that Calm-deficient (Calm−/−) mouse fibroblasts derived from fit1 mice are iron deficient, and are more sensitive to treatment with iron chelators. Objective: Since CALM-AF10 leukemia cells are haploinsufficient for CALM, we hypothesize that reduced levels of CALM in CALM-AF10 leukemia cells cause a relative iron deficiency, and result in enhanced sensitivity to iron deprivation. Design/Methods: Fibroblasts and fetal liver hematopoietic progenitors (HP) were derived from Calm−/−, Calm+/− or Calm+/+ E14 embryos. The proliferation rates of non-immortalized fibroblasts were compared in the presence and absence of supplemental iron (ferric ammonium citrate (FAC)) or treatment with an iron chelator (deferoxamine (DFO)). Surface transferrin receptor expression was quantified by flow cytometry. Primary HP cells were cultured in the presence or absence of DFO and viable cell numbers were determined. To examine the anti-leukemic effect of iron deprivation in vivo, C57BL/6J-Tyrc-2J mice were fed a low-iron diet, transplanted with CALM-AF10-transduced Calm+/− leukemia cells and leukemia latency was determined. Results: Heterozygous Calm+/− fibroblasts exhibit CALM protein levels that are intermediate between their wildtype (Calm+/+) and deficient (Calm−/−) counterparts. Calm+/− cells display a slower rate of proliferation in vitro (50% reduction of viable cells) compared to their wildtype fibroblasts, and this growth deficiency can be corrected by iron supplementation with 50 mM FAC. The presence of reduced intracellular iron levels in Calm+/− fibroblasts was manifested by increased transferrin receptor expression relative to wildtype cells. In vitro, Calm+/− HP cells showed greater sensitivity to iron chelation by DFO (5 mM) than Calm+/+ controls: the relative number of viable cells in the presence of DFO was 20% lower in Calm+/− compared to Calm+/− HP cells. To assess the impact of iron deprivation in vivo, bone marrow cells from Calm+/−CALM-AF10 leukemic mice were injected into secondary recipient mice preconditioned by having received a low-iron diet for 8 weeks (n=9). The onset of leukemia in transplanted mice maintained on the iron-deficient diet was considerably delayed (median survival 93 days versus 63 days, p=0.013) relative to age- and gender-matched control mice fed a normal diet (n=9). Conclusions: We have shown that reduced CALM expression impairs iron import and consequently limits the rate of cell proliferation. Both in vitro and in vivo results suggest that CALM haploinsufficient CALM-AF10 leukemias are particularly sensitive to iron deprivation. This raises the possibility that iron chelation may be a previously unappreciated treatment option for patients with aggressive CALM-rearranged leukemias. We are currently studying the impact of iron chelation in murine CALM-AF10 in vivo leukemia models. Disclosures: No relevant conflicts of interest to declare.

2007 ◽  
Vol 28 (4) ◽  
pp. 812-823 ◽  
Author(s):  
Richard Milner ◽  
Stephanie Hung ◽  
Xiaoyun Wang ◽  
Maria Spatz ◽  
Gregory J del Zoppo

During focal cerebral ischemia, the detachment of astrocytes from the microvascular basal lamina is not completely explained by known integrin receptor expression changes. Here, the impact of experimental ischemia (oxygen—glucose deprivation (OGD)) on dystroglycan expression by murine endothelial cells and astrocytes grown on vascular matrix laminin, perlecan, or collagen and the impact of middle cerebral artery occlusion on αβ-dystroglycan within cerebral microvessels of the nonhuman primate were examined. Dystroglycan was expressed on all cerebral microvessels in cortical gray and white matter, and the striatum. Astrocyte adhesion to basal lamina proteins was managed in part by α-dystroglycan, while ischemia significantly reduced expression of dystroglycan both in vivo and in vitro. Furthermore, dystroglycan and integrin α6β4 expressions on astrocyte end-feet decreased in parallel both in vivo and in vitro. The rapid loss of astrocyte dystroglycan during OGD appears protease-dependent, involving an matrix metalloproteinase-like activity. This may explain the rapid detachment of astrocytes from the microvascular basal lamina during ischemic injury, which could contribute to significant changes in microvascular integrity.


Endocrinology ◽  
2012 ◽  
Vol 153 (8) ◽  
pp. 3724-3734 ◽  
Author(s):  
María Celia Fernández ◽  
Marcela Venara ◽  
Susana Nowicki ◽  
Héctor E. Chemes ◽  
Marta Barontini ◽  
...  

IGFs are involved in malignant transformation and growth of several tissues, including the adrenal medulla. The present study was designed to evaluate the impact of IGF-I on pheochromocytoma development. We used a murine pheochromocytoma (MPC) cell line (MPC4/30) and an animal model with a reduction of 75% in circulating IGF-I levels [liver-IGF-I-deficient (LID) mice] to perform studies in vitro and in vivo. We found that, in culture, IGF-I stimulation increases proliferation, migration, and anchorage-independent growth, whereas it inhibits apoptosis of MPC cells. When injected to control and to LID mice, MPC cells grow and form tumors with features of pheochromocytoma. Six weeks after cell inoculation, all control mice developed sc tumors. In contrast, in 73% of LID mice, tumor development was delayed to 7–12 wk, and the remaining 27% did not develop tumors up to 12 wk after inoculation. LID mice harboring MPC cells and treated with recombinant human IGF-I (LID+) developed tumors as controls. Tumors developed in control, LID, and LID+ mice had similar histology and were similarly positive for IGF-I receptor expression. The apoptotic index was higher in tumors from LID mice compared with those from control mice, whereas vascular density was decreased. In summary, our work demonstrates that IGF-I has a critical role in maintaining tumor phenotype and survival of already transformed pheochromocytoma cells and is required for the initial establishment of these tumors, providing encouragement to carry on research studies to address the IGF-I/IGF-I receptor system as a target of therapeutic strategies for pheochromocytoma treatment in the future.


Neurosurgery ◽  
1995 ◽  
Vol 36 (6) ◽  
pp. 1158-1164 ◽  
Author(s):  
Dennis Y. Wen ◽  
Walter A. Hall ◽  
John Conrad ◽  
Aslak Godal ◽  
Vivi A. Flørenes ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3440-3440
Author(s):  
Danelle James ◽  
Esther D Avery ◽  
Ling Zhang ◽  
Jamie Gould ◽  
Andrew Greaves ◽  
...  

Abstract Abstract 3440 Poster Board III-328 Lenalidomide (L), an oral immunomodulatory agent used for the treatment of MM and MDS and has clinical activity in patients (pts) with relapsed/refractory CLL. When administered to CLL pts L is often associated with a TFR. TFR is painful enlargement of the lymph nodes and/or spleen. The mechanism of TFR is unknown. Previous reports have demonstrated upregulation of costimulatory molecules on leukemia cells with in-vitro L exposure. This led to the hypothesis that B cell activation is responsible for the TFR, possibly allowing for greater immune responses and infiltration of the lymph nodes/ spleen with inflammatory cells. Another hypothesis is that TFR results from the modulation of chemokine receptors allowing malignant lymphocytes to experience greater homing to the lymph nodes. We designed a phase II study to evaluate the efficacy and tolerability of L and rituximab as initial therapy of CLL. This previously untreated pt population provides a fairly homogenous population to assess the impact of in-vivo L treatment on CLL cells and test these hypotheses. To assess the impact of L on the CLL cells we designed a 3 week L run-in and performed ex-vivo analysis of the CLL cells prior to rituximab. All pts received L 2.5mg daily for the first 7 days and were escalated to 5 mg if they lacked significant toxicity on day 8. 20 pts have been enrolled on study and received at least 3 weeks of L. We report the early results of corollary science phenotyping the leukemia cells of 12 pts through the relative expression of a number of surface markers prior to and 21 days post initiation L. 9 of these pts experienced the TFR and 3 did not. 7 pts were on 5mg of L on day 21 and 5 on 2.5mg. Leukemia cells were stained for CD19, CD20 and chemokine receptors CXCR4, CXCR5, and CCR7 and for costimulatory molecules CD40, CD80, CD86, and CD54. Mean fluorescent intensity (MFI) was determined on CD19+ lymphocytes. MFI ratio (MFIR) was calculated by dividing the MFI for each stain by the isotype control of the respective sample. Two-sided student t test were used to assess statistical significance. Increased expression of CD40 was observed in 9/12 pts, (MFIR p 0.09) including 7/9 pts with TFR. Mean fold increase of CD40 MFIR for those with TFR was 1.13 compared with 1.26 in those without TFR (p = NS). CD86 increased in 6/12 including all 3 pts who did not experience TFR. Mean fold increase of CD86 MFIR for those with TFR was 1.11 and 1.6 in those without TFR (p = NS). CD80 MFIR increased slightly above pretreatment levels in 3/12 pts (3/9 with TFR). CD54 (ICAM) was evaluated in 9 pts and increased in 6. Median MFIR of CD54 following in vivo L increased from 14.7 to 24.8 (p = 0.066). Mean fold increase of CD54 MFIR for 2 pts without TFR was 2.08 compared with 1.82 for those with TFR (p= NS). MFIR for CXCR4 decreased in 10/12 pts during L including 8/9 pts with TFR. Median MFIR for CXCR4 prior to treatment was 67.9 and decreased to 37.5 (p = 0.024). Pts who experienced TFR had a more pronounced decrease in CXCR4 expression with MFIRs following L treatment mean 56% of pretreatment levels compared to the pts without TFR at 72% (p = NS). To assess if decreased CXCR4 expression could be the result of CLL cells mobilized from microenvironment niches we assessed the density of CXCR4 on 4 pts. All 4 pts had an increase in the population of CD19+ cells with dim CXCR4 expression from 10.2% prior to therapy to 26.4% following L (p = 0.058). MFIR for CXCR5 decreased in 9/12 pts (7/9 with TFR). For the 12 pts median CXCR5 MFIR 122.8 prior to treatment and 75.3 following 21 days of L (p = 0.045). Median MFIR for those who did or did not experience TFR was 79% and 80% of pretreatment levels respectively. There was not a significant change in CCR7 for the 12 pts or clear relationship to TFR. Previous studies reported in vitro downmodulation of CD20 on CLL cells exposed to L. As this clinical study is evaluating the combination of L and rituximab, we assessed CD20 levels on CLL cells exposed to L in vivo. Median MFIR for CD20 prior to treatment was 10.5 and was 8.5 following 21 days of L (p = 0.064). In the 12 pts evaluated CD20 expression increased in 5 pts (mean increase 24% (range 5-58%)) and decreased in the other 7 (mean decrease 31% (range 5-42%)). All pts clinically responded to the addition of rituximab with decrease in absolute lymphocyte count. These data show no clear relationship to the upregulation of markers of B cell activation on circulating CLL cells and the occurrence of TFR. There is early evidence of in-vivo down modulation of chemokine receptors CXCR4 and CXCR5, however this phenomenon is not markedly different in pts who have experienced TFR. Disclosures Off Label Use: Lenalidomide is not approved for the treatment of CLL.


Neurosurgery ◽  
1995 ◽  
Vol 36 (6) ◽  
pp. 1158???1164
Author(s):  
Dennis Y. Wen ◽  
Walter A. Hall ◽  
John Conrad ◽  
Aslak Godal ◽  
Vivi A. Fl??renes ◽  
...  

2019 ◽  
Author(s):  
Leslie M. Jonart ◽  
Maryam Ebadi ◽  
Patrick Basile ◽  
Kimberly Johnson ◽  
Jessica Makori ◽  
...  

AbstractProtection from acute lymphoblastic leukemia (ALL) relapse in the central nervous system (CNS) is crucial to survival and quality of life for ALL patients. Current CNS-directed therapies cause significant toxicities and are only partially effective. Moreover, the impact of the CNS microenvironment on leukemia biology is poorly understood. Herein, we showed that leukemia cells associated with the meninges of xenotransplanted mice, or co-cultured with meningeal cells, exhibit enhanced chemoresistance due to effects on both apoptosis balance and quiescence. From a mechanistic standpoint, we identified that leukemia chemoresistance is primarily mediated by direct leukemia-meningeal cell interactions and overcome by detaching the leukemia cells from the meninges. Next, we used a co-culture adhesion assay to identify drugs that disrupted leukemia-meningeal adhesion. In addition to identifying several drugs that inhibit canonical cell adhesion targets we found that Me6TREN, a novel hematopoietic stem cell (HSC) mobilizing compound, also disrupts leukemia-meningeal adhesion in vitro and in vivo. Finally, Me6TREN enhanced the efficacy of cytarabine in treating CNS leukemia in xenotransplanted mice. This work demonstrates that the meninges exert a critical influence on leukemia chemoresistance, elucidates mechanisms of CNS relapse beyond the well-described role of the blood-brain barrier, and identifies novel therapeutic approaches for overcoming chemoresistance.


2013 ◽  
Vol 150 (3) ◽  
pp. 1024-1031 ◽  
Author(s):  
Mohammad Hossein Boskabady ◽  
Sakine Shahmohammadi Mehrjardi ◽  
Abadorrahim Rezaee ◽  
Houshang Rafatpanah ◽  
Sediqeh Jalali

2021 ◽  
Vol 22 (5) ◽  
pp. 2530
Author(s):  
Bijean D. Ford ◽  
Diego Moncada Giraldo ◽  
Camilla Margaroli ◽  
Vincent D. Giacalone ◽  
Milton R. Brown ◽  
...  

Cystic fibrosis (CF) lung disease is dominated by the recruitment of myeloid cells (neutrophils and monocytes) from the blood which fail to clear the lung of colonizing microbes. In prior in vitro studies, we showed that blood neutrophils migrated through the well-differentiated lung epithelium into the CF airway fluid supernatant (ASN) mimic the dysfunction of CF airway neutrophils in vivo, including decreased bactericidal activity despite an increased metabolism. Here, we hypothesized that, in a similar manner to neutrophils, blood monocytes undergo significant adaptations upon recruitment to CFASN. To test this hypothesis, primary human blood monocytes were transmigrated in our in vitro model into the ASN from healthy control (HC) or CF subjects to mimic in vivo recruitment to normal or CF airways, respectively. Surface phenotype, metabolic and bacterial killing activities, and transcriptomic profile by RNA sequencing were quantified post-transmigration. Unlike neutrophils, monocytes were not metabolically activated, nor did they show broad differences in activation and scavenger receptor expression upon recruitment to the CFASN compared to HCASN. However, monocytes recruited to CFASN showed decreased bactericidal activity. RNASeq analysis showed strong effects of transmigration on monocyte RNA profile, with differences between CFASN and HCASN conditions, notably in immune signaling, including lower expression in the former of the antimicrobial factor ISG15, defensin-like chemokine CXCL11, and nitric oxide-producing enzyme NOS3. While monocytes undergo qualitatively different adaptations from those seen in neutrophils upon recruitment to the CF airway microenvironment, their bactericidal activity is also dysregulated, which could explain why they also fail to protect CF airways from infection.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Hiroaki Kanzaki ◽  
Tetsuhiro Chiba ◽  
Junjie Ao ◽  
Keisuke Koroki ◽  
Kengo Kanayama ◽  
...  

AbstractFGF19/FGFR4 autocrine signaling is one of the main targets for multi-kinase inhibitors (MKIs). However, the molecular mechanisms underlying FGF19/FGFR4 signaling in the antitumor effects to MKIs in hepatocellular carcinoma (HCC) remain unclear. In this study, the impact of FGFR4/ERK signaling inhibition on HCC following MKI treatment was analyzed in vitro and in vivo assays. Serum FGF19 in HCC patients treated using MKIs, such as sorafenib (n = 173) and lenvatinib (n = 40), was measured by enzyme-linked immunosorbent assay. Lenvatinib strongly inhibited the phosphorylation of FRS2 and ERK, the downstream signaling molecules of FGFR4, compared with sorafenib and regorafenib. Additional use of a selective FGFR4 inhibitor with sorafenib further suppressed FGFR4/ERK signaling and synergistically inhibited HCC cell growth in culture and xenograft subcutaneous tumors. Although serum FGF19high (n = 68) patients treated using sorafenib exhibited a significantly shorter progression-free survival and overall survival than FGF19low (n = 105) patients, there were no significant differences between FGF19high (n = 21) and FGF19low (n = 19) patients treated using lenvatinib. In conclusion, robust inhibition of FGF19/FGFR4 is of importance for the exertion of antitumor effects of MKIs. Serum FGF19 levels may function as a predictive marker for drug response and survival in HCC patients treated using sorafenib.


Sign in / Sign up

Export Citation Format

Share Document